Skip to main content
Erschienen in: Seminars in Immunopathology 4/2022

Open Access 01.07.2022 | Introduction

Tolerance and autoimmunity in the liver

verfasst von: Christoph Schramm, Ye H. Oo, Ansgar W. Lohse

Erschienen in: Seminars in Immunopathology | Ausgabe 4/2022

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN
Hinweise
This article is a contribution to the special issue on: Tolerance and autoimmunity in the liver - Guest Editors: Christoph Schramm, Ansgar Lohse & Ye Oo

Publisher's note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Autoimmune diseases arise from the failure of immune tolerance towards self-antigens. In the liver, maintenance of immune tolerance is particularly challenging. The liver filters all blood from the intestinal tract and is thus exposed to an infinite number of nutritional and microbial antigens. Additionally, the liver harbours the large mucosal surface of the biliary system which is colonized with microbiota and exposed to environmental factors. Together with neo-antigens arising from hepatic metabolism, the liver therefore is constantly challenged to maintain tolerance against harmless antigens and to rise immune responses against harmful environmental exposure. It is presumably for this reason that the liver microenvironment—under normal circumstances—is a particularly potent inducer of immune tolerance within the organ itself, but also systemically. For example, targeting liver sinusoidal endothelial cells using nanoparticles revealed that these cells can be harnessed to induce tolerance to the central nervous system autoantigen myelin basic protein, which can prevent the development of experimental autoimmune encephalomyelitis [1]. While the tolerogenic liver microenvironment prevents immune attack in the healthy liver, it presumably also provides a survival benefit for cancer cells and hepatotropic viruses by preventing their elimination by the immune system.
In spite of its tolerogenic potential, the liver can fall victim to three major autoimmune diseases for which curative therapies are lacking: autoimmune hepatitis (AIH), primary biliary cholangitis (PBC) and primary sclerosing cholangitis (PSC). Therefore, these diseases are leading indications for liver transplantation. Understanding the making and breaking of liver tolerance will enable the development of novel and effective immunotherapies. These insights are not only relevant for the treatment of the autoimmune liver diseases, as understanding these mechanisms can be key to develop tools to manipulate systemic immunity and to treat autoimmune diseases outside of the liver and bile ducts.
Within this special issue of Seminars in Immunopathology, we highlight recent advances in the understanding of mechanisms governing tolerance and autoimmunity in the liver. We focus on research areas with a clear potential for novel therapeutic strategies. Genetic risk and environmental exposures both contribute to the development of autoimmunity in the liver. Among the autoimmune liver diseases, genetic risk is highest for PBC. Genome-wide association studies have highlighted the potential contribution of the immune system, and specifically the adaptive immune system for disease pathogenesis. However, functional studies on polymorphisms associated with disease risk are largely lacking. Summarizing how genetic risk contributes to autoimmune liver disease, Ellinghaus gives an outlook on how genetic analyses can improve our understanding of disease mechanisms in the future. This also includes the identification of autoantigens, information on which is largely lacking [2]. Autoantibodies are characteristic of AIH and PBC and have recently been associated with PSC prognosis as well. Additionally, AIH is characterized by selective elevation of serum Immunoglobulin G levels and by infiltration of the liver by B cells and plasma cells. Few case series suggested that B cell depletion therapy may improve disease activity in difficult to treat AIH patients. Schultheiß and colleagues discuss whether B cells in AIH are only bystanders or central players of disease pathogenesis and thus therapeutic targets in AIH [3]. Mucosa-associated invariant T (MAIT) cells are highly abundant in human liver. Their invariant T cell receptor recognizes bacterial metabolites of B vitamins. MAIT cells also respond to a number of cytokines expressed in liver inflammation. It can be suggested that MAIT cells play an important role at the mucosal barrier of bile ducts, acquiring either regenerative or pro-inflammatory and pro-fibrogenic potential according to the environmental exposure. Mehta et al. summarize the current knowledge on MAIT cells in the liver and how different ways of stimulation may shape their function within human liver [4]. Targeting cytokines may be an attractive therapeutic option independent from specific cell types. TNF is a central pro-inflammatory cytokine in autoimmune diseases including AIH. It has recently been shown that neutralizing TNF can be used to effectively control difficult to treat AIH patients. On the other hand, anti-TNF treatment can induce liver inflammation in patients with extrahepatic immune-mediated diseases such as inflammatory bowel disease. It is therefore essential to dissect the role of TNF in immune homeostasis and autoimmunity in the liver in order to develop precise and well-tolerated therapies. Tiegs et al. summarize current knowledge on TNF, signaling through TNF receptors I and II and how this could lead to novel therapies of autoimmune liver diseases [5]. Regulatory T cells (Treg) belong to the body’s armamentarium to maintain and restore peripheral immune tolerance, both in an antigen-specific and -unspecific way. Targeting Treg numbers and function currently is intensely investigated as therapy for many autoimmune diseases. Deficiencies of Treg have been reported in autoimmune liver diseases, but remain a matter of debate depending on disease and tissue analyzed. Oo and colleagues have recently reported the migration of ex vivo expanded Treg into the liver of people with AIH [6] and here review the challenges and opportunities in achieving effective Treg therapy in autoimmune liver diseases as a promising future tool to improve treatment without harmful side effects [7].
As mentioned above, the liver’s tolerogenic potential can be strengthened to treat hepatic autoimmunity, but it may also be exploited to induce systemic immune tolerance. One of the hepatic cell types with a high tolerogenic potential are the hepatic sinusoidal endothelial cells, that form the lining of liver sinusoids and are thus exposed first line to environmental factors entering the liver via the portal vein and thus the intestinal tract. Gottwick and colleagues therefore focus in their review on how the liver can be harnessed to induce antigen-specific immune tolerance systemically [8]. Microbial products and microbiota themselves enter the liver via the portal vein. In addition, the portal tracts of the liver are constantly exposed to the luminal and mucosal microbiota present in the biliary system. It is now clear that autoimmune liver disease patients harbour a microbiota composition that is different from healthy people and different between the diseases. Additionally, it has recently been shown that bile fluid harbours a rich microbiota composition that is different in people with PSC [9]. In this special issue, Liwinski et al. amend this concept and highlight current evidence and concepts on how the intestinal and biliary microbiome affect autoimmune liver disease [10]. Along this line, Nakamoto et al. comprehensively review the current understanding of IL-17 producing CD4 + T cells (Th17) in autoimmune liver diseases and specifically in PSC. IL-17 plays an important role in pathogen defence, e.g. by recruiting neutrophil granulocytes. On the other hand, IL-17 has emerged as a promising treatment target in several autoimmune diseases outside of the liver. Thus, Nakamoto and colleagues have recently described a pathogenetic role of microbiota-induced Th17 cells in PSC pathogenesis [11]. Here, they now review the current understanding of the dual role of IL-17 and Th17 cells in balancing autoimmunity and pathogen defence [12]. Biliary epithelial cells form the mucosal barrier of bile ducts and are targets of autoimmune inflammation in PBC and PSC. They may not only be targets, but also active contributors to liver inflammation. Their senescence-associated secretory phenotype has been shown to promote periductal inflammation. Trussoni et al. summarize current understanding of cellular senescence and its role as a driver of immunopathology and therapeutic target in the cholangiopathies [13]. To conclude this special issue, Bertolini and colleagues review the central role of bile acids and their receptors as modulators and therapeutic targets in liver inflammation [14]. Indeed, bile acid derivatives are the only licenced treatment options for PBC and drugs targeting the Farneoid X Receptor (FXR), the transcription factor central to the regulation of bile acid homeostasis, are being studied for the treatment not only of PBC and PSC, but also for other liver diseases such as non-alcoholic (or metabolic associated) steatohepatitis. Additionally, the uptake of bile acids is targeted for treatment of hereditary cholestatic liver diseases, underlining the important role bile acids play in immune mediated and autoimmune liver diseases.
Taken together, in this special issue, we highlight some of the topics important for the development of future therapies for autoimmune liver diseases. Some of these concepts have started to enter the clinic and care of patients, others are being developed to offer more targeted and better tolerated therapies for patients with autoimmune liver diseases. Current treatment, if available, often is associated with severe side effects, ineffective in a considerable proportion of patients and unable to induce cure. With increasing knowledge of the mechanisms underlying tolerance and autoimmunity in the liver, there is realistic hope to improve the care of these difficult to treat diseases in the future.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
download
DOWNLOAD
print
DRUCKEN

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Carambia A, Freund B, Schwinge D, Bruns OT, Salmen SC, Ittrich H, Reimer R, Heine M, Huber S, Waurisch C, Eychmüller A, Wraith DC, Korn T, Nielsen P, Weller H, Schramm C, Lüth S, Lohse AW, Heeren J, Herkel J (2015) Nanoparticle-based autoantigen delivery to Treg-inducing liver sinusoidal endothelial cells enables control of autoimmunity in mice. J Hepatol 62(6):1349–1356CrossRef Carambia A, Freund B, Schwinge D, Bruns OT, Salmen SC, Ittrich H, Reimer R, Heine M, Huber S, Waurisch C, Eychmüller A, Wraith DC, Korn T, Nielsen P, Weller H, Schramm C, Lüth S, Lohse AW, Heeren J, Herkel J (2015) Nanoparticle-based autoantigen delivery to Treg-inducing liver sinusoidal endothelial cells enables control of autoimmunity in mice. J Hepatol 62(6):1349–1356CrossRef
6.
Zurück zum Zitat Oo YH, Ackrill S, Cole R, Jenkins L, Anderson P, Jeffery HC, Jones N, Jeffery LE, Lutz P, Wawman RE, Athwal AK, Thompson J, Gray J, Guo K, Barton D, Hirschfield GM, Wong T, Guest P, Adams DH (2019) Liver homing of clinical grade Tregs after therapeutic infusion in patients with autoimmune hepatitis. JHEP Rep 1(4):286–296CrossRef Oo YH, Ackrill S, Cole R, Jenkins L, Anderson P, Jeffery HC, Jones N, Jeffery LE, Lutz P, Wawman RE, Athwal AK, Thompson J, Gray J, Guo K, Barton D, Hirschfield GM, Wong T, Guest P, Adams DH (2019) Liver homing of clinical grade Tregs after therapeutic infusion in patients with autoimmune hepatitis. JHEP Rep 1(4):286–296CrossRef
9.
Zurück zum Zitat Liwinski T, Zenouzi R, John C, Ehlken H, Rühlemann MC, Bang C, Groth S, Lieb W, Kantowski M, Andersen N, Schachschal G, Karlsen TH, Hov JR, Rösch T, Lohse AW, Heeren J, Franke A, Schramm C (2020) Alterations of the bile microbiome in primary sclerosing cholangitis. Gut 69(4):665–672CrossRef Liwinski T, Zenouzi R, John C, Ehlken H, Rühlemann MC, Bang C, Groth S, Lieb W, Kantowski M, Andersen N, Schachschal G, Karlsen TH, Hov JR, Rösch T, Lohse AW, Heeren J, Franke A, Schramm C (2020) Alterations of the bile microbiome in primary sclerosing cholangitis. Gut 69(4):665–672CrossRef
11.
Zurück zum Zitat Nakamoto N, Sasaki N, Aoki R, Miyamoto K, Suda W, Teratani T, Suzuki T, Koda Y, Chu PS, Taniki N, Yamaguchi A, Kanamori M, Kamada N, Hattori M, Ashida H, Sakamoto M, Atarashi K, Narushima S, Yoshimura A, Honda K, Sato T, Kanai T (2019) Gut pathobionts underlie intestinal barrier dysfunction and liver T helper 17 cell immune response in primary sclerosing cholangitis. Nat Microbiol 4(3):492–503CrossRef Nakamoto N, Sasaki N, Aoki R, Miyamoto K, Suda W, Teratani T, Suzuki T, Koda Y, Chu PS, Taniki N, Yamaguchi A, Kanamori M, Kamada N, Hattori M, Ashida H, Sakamoto M, Atarashi K, Narushima S, Yoshimura A, Honda K, Sato T, Kanai T (2019) Gut pathobionts underlie intestinal barrier dysfunction and liver T helper 17 cell immune response in primary sclerosing cholangitis. Nat Microbiol 4(3):492–503CrossRef
Metadaten
Titel
Tolerance and autoimmunity in the liver
verfasst von
Christoph Schramm
Ye H. Oo
Ansgar W. Lohse
Publikationsdatum
01.07.2022
Verlag
Springer Berlin Heidelberg
Erschienen in
Seminars in Immunopathology / Ausgabe 4/2022
Print ISSN: 1863-2297
Elektronische ISSN: 1863-2300
DOI
https://doi.org/10.1007/s00281-022-00952-6

Weitere Artikel der Ausgabe 4/2022

Seminars in Immunopathology 4/2022 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Echinokokkose medikamentös behandeln oder operieren?

06.05.2024 DCK 2024 Kongressbericht

Die Therapie von Echinokokkosen sollte immer in spezialisierten Zentren erfolgen. Eine symptomlose Echinokokkose kann – egal ob von Hunde- oder Fuchsbandwurm ausgelöst – konservativ erfolgen. Wenn eine Op. nötig ist, kann es sinnvoll sein, vorher Zysten zu leeren und zu desinfizieren. 

Wo hapert es noch bei der Umsetzung der POMGAT-Leitlinie?

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

Das Risiko für Vorhofflimmern in der Bevölkerung steigt

02.05.2024 Vorhofflimmern Nachrichten

Das Risiko, im Lauf des Lebens an Vorhofflimmern zu erkranken, ist in den vergangenen 20 Jahren gestiegen: Laut dänischen Zahlen wird es drei von zehn Personen treffen. Das hat Folgen weit über die Schlaganfallgefährdung hinaus.

VHF-Ablation nützt wohl nur bei reduzierter Auswurfleistung

02.05.2024 Ablationstherapie Nachrichten

Ob die Katheterablation von Vorhofflimmern bei Patienten mit Herzinsuffizienz die Komplikationsraten senkt, scheint davon abzuhängen, ob die Auswurfleistung erhalten ist oder nicht. Das legen die Ergebnisse einer Metaanalyse nahe.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.