1
|
Liu J, Wang H, Zhang S, Liu J. Identification of shared and disease-specific intratumoral microbiome-host gene associations in gastrointestinal tumors. Physiol Genomics 2024; 56:699-710. [PMID: 39250148 DOI: 10.1152/physiolgenomics.00036.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/24/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024] Open
Abstract
Intratumoral microbiota and host genes interact to promote gastrointestinal disorders, but how the two interact to influence host tumorigenesis remains unclear. Here, we utilized a machine learning-based framework to jointly dissect the paired intratumoral microbiome and host transcriptome profiles in patients with colon adenocarcinoma, hepatocellular carcinoma, and gastric cancer. We identified associations between intratumoral microbes and host genes that depict shared as well as cancer type-specific patterns. We found that a common set of host genes and pathways implicated in cell proliferation and energy metabolism are associated with cancer type-specific intratumoral microbes. In addition, we also found that intratumoral microbes that have been implicated in three gastrointestinal tumors, such as Lachnoclostridium, are correlated with different host pathways in each tumor, indicating that similar microbes can influence host tumorigenesis in a cancer type-specific manner by regulation of different host genes. Our study reveals patterns of association between intratumoral microbiota and host genes in gastrointestinal tumors, providing new insights into the biology of gastrointestinal tumors.NEW & NOTEWORTHY Our study constitutes a pivotal advancement in elucidating the intricate relationship between the intratumoral microbiome and host gene regulation, thereby gaining insights into the pivotal role that the intratumoral microbiome plays in the etiology of gastrointestinal tumors.
Collapse
Affiliation(s)
- Jing Liu
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Hongyan Wang
- Department of Pathology, Daqing Oilfield General Hospital, Daqing, People's Republic of China
| | - Shuai Zhang
- Department of Neurology, Shanghai Anda Hospital, Shanghai, People's Republic of China
| | - Jinyang Liu
- Geneis Beijing Company, Limited., Beijing, People's Republic of China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, People's Republic of China
| |
Collapse
|
2
|
Dorner H, Stolzer I, Mattner J, Kaminski S, Leistl S, Edrich LM, Schwendner R, Hobauer J, Sebald A, Leikam S, Gonzalez Acera M, Düll M, Lang R, Seidel G, Seitz T, Hellerbrand C, Fuhrmann G, Distler U, Tenzer S, Eichhorn P, Vieth M, Schramm C, Arnold P, Becker C, Weidinger C, Siegmund B, Atreya R, Leppkes M, Naschberger E, Sampaziotis F, Dietrich P, Rauh M, Wirtz S, Kremer AE, Neurath MF, Günther C. Gut Pathobiont-Derived Outer Membrane Vesicles Drive Liver Inflammation and Fibrosis in Primary Sclerosing Cholangitis-Associated Inflammatory Bowel Disease. Gastroenterology 2024; 167:1183-1197.e16. [PMID: 38992449 DOI: 10.1053/j.gastro.2024.06.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/13/2024]
Abstract
BACKGROUND & AIMS Primary sclerosing cholangitis (PSC), often associated with inflammatory bowel disease (IBD), presents a multifactorial etiology involving genetic, immunologic, and environmental factors. Gut dysbiosis and bacterial translocation have been implicated in PSC-IBD, yet the precise mechanisms underlying their pathogenesis remain elusive. Here, we describe the role of gut pathobionts in promoting liver inflammation and fibrosis due to the release of bacterial outer membrane vesicles (OMVs). METHODS Preclinical mouse models in addition to ductal organoids were used to acquire mechanistic data. A proof-of-concept study including serum and liver biopsies of a patient cohort of PSC (n = 22), PSC-IBD (n = 45), and control individuals (n = 27) was performed to detect OMVs in the systemic circulation and liver. RESULTS In both preclinical model systems and in patients with PSC-IBD, the translocation of OMVs to the liver correlated with enhanced bacterial sensing and accumulation of the NLRP3 inflammasome. Using ductal organoids, we were able to precisely attribute the pro-inflammatory and pro-fibrogenic properties of OMVs to signaling pathways dependent on Toll-like receptor 4 and NLRP3-gasdermin-D. The immunostimulatory potential of OMVs could be confirmed in macrophages and hepatic stellate cells. Furthermore, when we administered gut pathobiont-derived OMVs to Mdr2-/- mice, we observed a significant enhancement in liver inflammation and fibrosis. In a translational approach, we substantiated the presence of OMVs in the systemic circulation and hepatic regions of severe fibrosis using a PSC-IBD patient cohort. CONCLUSIONS This study demonstrates the contribution of gut pathobionts in releasing OMVs that traverse the mucosal barrier and, thus, promote liver inflammation and fibrosis in PSC-IBD. OMVs might represent a critical new environmental factor that interacts with other disease factors to cause inflammation and thus define potential new targets for fibrosis therapy.
Collapse
Affiliation(s)
- Heidrun Dorner
- Department of Medicine 1, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Iris Stolzer
- Department of Medicine 1, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Jochen Mattner
- Institute of Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Friedrich-Alexander-Universität Erlangen-Nürnberg Profile Center Immunomedicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sophie Kaminski
- Department of Medicine 1, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sofia Leistl
- Department of Medicine 1, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Lisa-Maria Edrich
- Department of Medicine 1, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Raphael Schwendner
- Department of Medicine 1, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Julia Hobauer
- Department of Medicine 1, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Adrian Sebald
- Department of Medicine 1, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Stefanie Leikam
- Department of Medicine 1, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Miguel Gonzalez Acera
- Department of Medicine 1, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Miriam Düll
- Department of Medicine 1, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Roland Lang
- Institute of Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Friedrich-Alexander-Universität Erlangen-Nürnberg Profile Center Immunomedicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Gerald Seidel
- Microbiology, Department of Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Tatjana Seitz
- Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Claus Hellerbrand
- Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Gregor Fuhrmann
- Department of Biology, Pharmaceutical Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ute Distler
- Institute of Immunology, University Medical Center of the Johannes-Gutenberg University, Mainz, Germany
| | - Stefan Tenzer
- Institute of Immunology, University Medical Center of the Johannes-Gutenberg University, Mainz, Germany
| | - Phillip Eichhorn
- Institute of Pathology, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Michael Vieth
- Institute of Pathology, Klinikum Bayreuth, Friedrich-Alexander-Universität Erlangen-Nürnberg, Bayreuth, Germany
| | - Christoph Schramm
- Department of Medicine, Martin Zeitz Center for Rare Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Philipp Arnold
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christoph Becker
- Department of Medicine 1, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Friedrich-Alexander-Universität Erlangen-Nürnberg Profile Center Immunomedicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Carl Weidinger
- Division of Gastroenterology, Infectiology and Rheumatology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Britta Siegmund
- Division of Gastroenterology, Infectiology and Rheumatology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Raja Atreya
- Department of Medicine 1, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Friedrich-Alexander-Universität Erlangen-Nürnberg Profile Center Immunomedicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Moritz Leppkes
- Department of Medicine 1, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Elisabeth Naschberger
- Division of Molecular and Experimental Surgery, Department of Surgery, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Fotios Sampaziotis
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge, United Kingdom; Cambridge Liver Unit, Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, United Kingdom; Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Peter Dietrich
- Department of Medicine 1, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Manfred Rauh
- Research Laboratory, Division of Pediatrics, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Stefan Wirtz
- Department of Medicine 1, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Friedrich-Alexander-Universität Erlangen-Nürnberg Profile Center Immunomedicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Andreas E Kremer
- Department of Medicine 1, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie, Universitätsklinikum Erlangen, Erlangen, Germany; Department of Gastroenterology and Hepatology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Markus F Neurath
- Department of Medicine 1, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Friedrich-Alexander-Universität Erlangen-Nürnberg Profile Center Immunomedicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Claudia Günther
- Department of Medicine 1, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Friedrich-Alexander-Universität Erlangen-Nürnberg Profile Center Immunomedicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie, Universitätsklinikum Erlangen, Erlangen, Germany.
| |
Collapse
|
3
|
Tang Z, Zhong M, Cao H, Wang Y, Guan G, Wang G, Wu J, Han F, Gao Y, Zhang K. Polysaccharide of Dicliptera chinensis (L.) Juss. alleviated cholestatic liver disease by modulating the FXR pathway. Int J Biol Macromol 2024; 281:136393. [PMID: 39383897 DOI: 10.1016/j.ijbiomac.2024.136393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/23/2024] [Accepted: 10/05/2024] [Indexed: 10/11/2024]
Abstract
Dicliptera chinensis (L.) Juss., is an herb known for its anti-inflammatory and anti-oxidant properties. In the previous studies, the chemical composition of the polysaccharide from Dicliptera chinensis (L.) Juss. (DCP) has been characterized as consisting of DCP1 and DCP2, of which DCP2 has hepatoprotective effects. The study examined the hepatoprotective potential of DCP2 against alpha-naphthyl isothiocyanate (ANIT)-induced cholestatic liver disease (CLD). In this study, RNA sequencing identified key research pathways involving bile acid metabolism, oxidative stress, and inflammation. Furthermore, qRT-PCR and Western blot analyses were conducted to further characterize these pathways. Additionally, the study included in vitro experiments with HepG2 cells to further investigate the effects of DCP2 on bile acid metabolism. In summary, the protective effect of DCP2 on the liver was reflected in alleviating the inflammatory response and oxidative stress, regulating the metabolism of bile acids, and mitigating liver damage caused by bile acids. This study further elucidated the hepatoprotective effects of DCP2 by examining its ability to counteract ANIT-induced CLD, suggesting that DCP2 is a promising biomacromolecule for hepatoprotection.
Collapse
Affiliation(s)
- Zixuan Tang
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, China; Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China
| | - Mingli Zhong
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, China
| | - Houkang Cao
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, China
| | - Yongwang Wang
- Department of Anesthesiology, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Guoqiang Guan
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, China
| | - Gang Wang
- Guangxi Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, Guilin 541004, China
| | - Jianzhao Wu
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, China
| | - Fei Han
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, China
| | - Ya Gao
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, China; Guangxi Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, Guilin 541004, China.
| | - Kefeng Zhang
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, China; Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China.
| |
Collapse
|
4
|
Fuchs CD, Simbrunner B, Baumgartner M, Campell C, Reiberger T, Trauner M. Bile acid metabolism and signaling in liver disease. J Hepatol 2024:S0168-8278(24)02572-8. [PMID: 39349254 DOI: 10.1016/j.jhep.2024.09.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 09/12/2024] [Accepted: 09/17/2024] [Indexed: 10/02/2024]
Abstract
Bile acids (BAs) have signaling functions efficiently regulating their own metabolism and transport as well as key aspects of lipid and glucose homeostasis. BAs shape the gut microbial flora and conversely are metabolized by microbiota. Disruption of BA transport, metabolism and physiological signaling function contribute to the pathogenesis and progression of a wide range of liver diseases including cholestatic disorders and metabolic dysfunction-associated steatotic liver disease (MASLD) as well as hepatocellular and cholangiocellular carcinoma. Additionally, impaired BA signaling may also affect the intestine and kidney, thereby contributing to failure of gut integrity driving the progression and complications of portal hypertension, cholemic nephropathy and the development of extrahepatic malignancies such as colorectal cancer. This review will summarize recent advances in the understanding of BA signaling, metabolism and transport focusing on transcriptional regulation and novel BA-focused therapeutic strategies for cholestatic and metabolic liver diseases.
Collapse
Affiliation(s)
- Claudia D Fuchs
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Benedikt Simbrunner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Maximillian Baumgartner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Clarissa Campell
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Thomas Reiberger
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
5
|
Ziegler A, Sæves I, Almaas R. Differences in bile acid profiles between cholestatic diseases - Development of a high throughput assay for dried bloodspots. Clin Chim Acta 2024; 562:119864. [PMID: 38992821 DOI: 10.1016/j.cca.2024.119864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/13/2024]
Abstract
BACKGROUND Cholestasis causes accumulation of bile acids (BAs) and changes the circulating bile acid profile. Quantification of circulating BAs in dried bloodspots (DBS) may demonstrate obstruction of bile flow and altered bile acid metabolism in the liver. High sample throughput enables rapid screening of cholestatic diseases. MATERIALS AND METHODS Ultra high performance liquid chromatography coupled to tandem mass spectrometry (UHPLC-MS/MS) was used for optimizing separation and detection of the primary unconjugated BAs cholic acid (CA) and chenodeoxycholic acid (CDCA); the secondary unconjugated BAs ursodeoxycholic acid (UDCA), hyodeoxycholic acid (HDCA) and deoxycholic acid (DCA), as well as the glycine- and taurine-conjugated variants of CA, CDCA, DCA and UDCA. Donor blood was obtained to prepare DBS calibrators and quality controls for method development and validation. RESULTS We developed a quantitative bile acid assay with a run-time of two minutes, and one-step sample preparation of 3.2 mm DBS discs. Validation results demonstrated overall good performance and was considered fit for purpose. Children with Alagille syndrome, Aagenaes syndrome and alpha-1 antitrypsin deficiency had increased BAs in DBS from newborn screening samples compared with age matched controls, and had different bile acids profiles. CONCLUSION We propose that our high throughput assay allows bile acid profiling in DBS that can be a valuable assessment tool for early screening of cholestasis in children. Assaying BAs in dried bloodspots is key for early detection of cholestasis, and provides transferability to a newborn screening setting.
Collapse
Affiliation(s)
- Anders Ziegler
- Department of Pediatric Research, Oslo University Hospital, Sognsvannsveien 20, 0372 Oslo, Norway; Department of Newborn Screening, Oslo University Hospital, Forskningsveien 2B, 0373 Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
| | - Ingjerd Sæves
- Department of Newborn Screening, Oslo University Hospital, Forskningsveien 2B, 0373 Oslo, Norway
| | - Runar Almaas
- Department of Pediatric Research, Oslo University Hospital, Sognsvannsveien 20, 0372 Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
6
|
Jyotsna, Sarkar B, Yadav M, Deka A, Markandey M, Sanyal P, Nagarajan P, Gaikward N, Ahuja V, Mohanty D, Basak S, Gokhale RS. A hepatocyte-specific transcriptional program driven by Rela and Stat3 exacerbates experimental colitis in mice by modulating bile synthesis. eLife 2024; 12:RP93273. [PMID: 39137024 PMCID: PMC11321761 DOI: 10.7554/elife.93273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024] Open
Abstract
Hepatic factors secreted by the liver promote homeostasis and are pivotal for maintaining the liver-gut axis. Bile acid metabolism is one such example wherein, bile acid synthesis occurs in the liver and its biotransformation happens in the intestine. Dysfunctional interactions between the liver and the intestine stimulate varied pathological outcomes through its bidirectional portal communication. Indeed, aberrant bile acid metabolism has been reported in inflammatory bowel disease (IBD). However, the molecular mechanisms underlying these crosstalks that perpetuate intestinal permeability and inflammation remain obscure. Here, we identify a novel hepatic gene program regulated by Rela and Stat3 that accentuates the inflammation in an acute experimental colitis model. Hepatocyte-specific ablation of Rela and Stat3 reduces the levels of primary bile acids in both the liver and the gut and shows a restricted colitogenic phenotype. On supplementation of chenodeoxycholic acid (CDCA), knock-out mice exhibit enhanced colitis-induced alterations. This study provides persuasive evidence for the development of multi-organ strategies for treating IBD and identifies a hepatocyte-specific Rela-Stat3 network as a promising therapeutic target.
Collapse
Affiliation(s)
- Jyotsna
- Immunometabolism Laboratory, National Institute of ImmunologyNew DelhiIndia
| | - Binayak Sarkar
- Immunometabolism Laboratory, National Institute of ImmunologyNew DelhiIndia
| | - Mohit Yadav
- Immunometabolism Laboratory, National Institute of ImmunologyNew DelhiIndia
| | - Alvina Deka
- System Immunology Laboratory, National Institute of ImmunologyNew DelhiIndia
| | - Manasvini Markandey
- Department of GastroEnterology, All India Institute of Medical SciencesNew DelhiIndia
| | | | - Perumal Nagarajan
- Immunometabolism Laboratory, National Institute of ImmunologyNew DelhiIndia
| | | | - Vineet Ahuja
- Department of GastroEnterology, All India Institute of Medical SciencesNew DelhiIndia
| | - Debasisa Mohanty
- Immunometabolism Laboratory, National Institute of ImmunologyNew DelhiIndia
| | - Soumen Basak
- System Immunology Laboratory, National Institute of ImmunologyNew DelhiIndia
| | - Rajesh S Gokhale
- Immunometabolism Laboratory, National Institute of ImmunologyNew DelhiIndia
- Department of Biology, Indian Institute of Science Education and ResearchPashanIndia
| |
Collapse
|
7
|
Hov JR. Sclerosing cholangitis and inflammatory bowel disease: a paradoxical relationship? Gut 2024:gutjnl-2024-332835. [PMID: 39025493 DOI: 10.1136/gutjnl-2024-332835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 07/10/2024] [Indexed: 07/20/2024]
Affiliation(s)
- Johannes R Hov
- Norwegian PSC Research Center and Section of Gastroenterology, Division of Surgery and Specialized Medicine, Oslo University Hospital, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
8
|
Bedke T, Stumme F, Tomczak M, Steglich B, Jia R, Bohmann S, Wittek A, Kempski J, Göke E, Böttcher M, Reher D, Franke A, Lennartz M, Clauditz T, Sauter G, Fründt T, Weidemann S, Tiegs G, Schramm C, Gagliani N, Pelczar P, Huber S. Protective function of sclerosing cholangitis on IBD. Gut 2024; 73:1292-1301. [PMID: 38839272 PMCID: PMC11287650 DOI: 10.1136/gutjnl-2023-330856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 05/09/2024] [Indexed: 06/07/2024]
Abstract
OBJECTIVE There is a strong clinical association between IBD and primary sclerosing cholangitis (PSC), a chronic disease of the liver characterised by biliary inflammation that leads to strictures and fibrosis. Approximately 60%-80% of people with PSC will also develop IBD (PSC-IBD). One hypothesis explaining this association would be that PSC drives IBD. Therefore, our aim was to test this hypothesis and to decipher the underlying mechanism. DESIGN Colitis severity was analysed in experimental mouse models of colitis and sclerosing cholangitis, and people with IBD and PSC-IBD. Foxp3+ Treg-cell infiltration was assessed by qPCR and flow cytometry. Microbiota profiling was carried out from faecal samples of people with IBD, PSC-IBD and mouse models recapitulating these diseases. Faecal microbiota samples collected from people with IBD and PSC-IBD were transplanted into germ-free mice followed by colitis induction. RESULTS We show that sclerosing cholangitis attenuated IBD in mouse models. Mechanistically, sclerosing cholangitis causes an altered intestinal microbiota composition, which promotes Foxp3+ Treg-cell expansion, and thereby protects against IBD. Accordingly, sclerosing cholangitis promotes IBD in the absence of Foxp3+ Treg cells. Furthermore, people with PSC-IBD have an increased Foxp3+ expression in the colon and an overall milder IBD severity. Finally, by transplanting faecal microbiota into gnotobiotic mice, we showed that the intestinal microbiota of people with PSC protects against colitis. CONCLUSION This study shows that PSC attenuates IBD and provides a comprehensive insight into the mechanisms involved in this effect.
Collapse
Affiliation(s)
- Tanja Bedke
- I. Department of Medicine, Section of Molecular Immunology and Gastroenterology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friederike Stumme
- I. Department of Medicine, Section of Molecular Immunology and Gastroenterology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Miriam Tomczak
- I. Department of Medicine, Section of Molecular Immunology and Gastroenterology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Babett Steglich
- I. Department of Medicine, Section of Molecular Immunology and Gastroenterology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Rongrong Jia
- I. Department of Medicine, Section of Molecular Immunology and Gastroenterology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Simon Bohmann
- I. Department of Medicine, Section of Molecular Immunology and Gastroenterology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Agnes Wittek
- I. Department of Medicine, Section of Molecular Immunology and Gastroenterology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Kempski
- I. Department of Medicine, Section of Molecular Immunology and Gastroenterology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Emilia Göke
- I. Department of Medicine, Section of Molecular Immunology and Gastroenterology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marius Böttcher
- I. Department of Medicine, Section of Molecular Immunology and Gastroenterology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dominik Reher
- I. Department of Medicine, Section of Molecular Immunology and Gastroenterology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anissa Franke
- I. Department of Medicine, Section of Molecular Immunology and Gastroenterology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maximilian Lennartz
- Center of Diagnostics, Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Pathology with the Section Molecular Pathology and Cytopathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Till Clauditz
- Center of Diagnostics, Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Pathology with the Section Molecular Pathology and Cytopathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Guido Sauter
- Center of Diagnostics, Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Pathology with the Section Molecular Pathology and Cytopathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorben Fründt
- I.Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sören Weidemann
- Center of Diagnostics, Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gisa Tiegs
- Center for Experimental Medicine, Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph Schramm
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- I.Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Martin Zeitz Center for Rare Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicola Gagliani
- I. Department of Medicine, Section of Molecular Immunology and Gastroenterology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Penelope Pelczar
- I. Department of Medicine, Section of Molecular Immunology and Gastroenterology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Samuel Huber
- I. Department of Medicine, Section of Molecular Immunology and Gastroenterology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
9
|
Fan Q, Liang R, Chen M, Li Z, Tao X, Ren H, Sheng Y, Li J, Lin R, Zhao C, She G. Metabolic characteristics of evodiamine were associated with its hepatotoxicity via PPAR/PI3K/AKT/NF-кB/tight junction pathway-mediated apoptosis in zebrafish. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 279:116448. [PMID: 38754199 DOI: 10.1016/j.ecoenv.2024.116448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/06/2024] [Accepted: 05/08/2024] [Indexed: 05/18/2024]
Abstract
Evodiae Fructus (EF), an herbal medicine, possesses remarkable anti-inflammatory and analgesic properties. It exhibits insecticidal activity as a potent insecticide candidate. However, the toxic characteristics of EF and the underlying mechanisms have not been comprehensively elucidated comprehensively. Thus, we comprehensively explored the toxic components of EF and established the relationship between the therapeutic and toxic effects of EF, encouraging its therapeutic use. We found that evodiamine (EVO), one of the main ingredients of EF, can truly reflect its analgesic properties. In phenotype observation trials, low doses of EVO (< 35 ng/mL) exhibited distinct analgesic activity without any adverse effects in zebrafish. However, EVO dose-dependently led to gross morphological abnormalities in the liver, followed by pericardial edema, and increased myocardial concentrations. Furthermore, the toxic effects of EVO decreased after processing in liver microsomes but increased after administering CYP450 inhibitors in zebrafish, highlighting the prominent effect of CYP450s in EVO-mediated hepatotoxicity. EVO significantly changed the expression of genes enriched in multiple pathways and biological processes, including lipid metabolism, inflammatory response, tight junction damage, and cell apoptosis. Importantly, the PPAR/PI3K/AKT/NF-кB/tight junction-mediated apoptosis pathway was confirmed as a critical functional signaling pathway inducing EVO-mediated hepatotoxicity. This study provided a typical example of the overall systematic evaluation of traditional Chinese medicine (TCM) and its active ingredients with significant therapeutic effects and simultaneous toxicities, especially metabolic toxicities.
Collapse
Affiliation(s)
- Qiqi Fan
- Beijing University of Chinese Medicine, Beijing 100102,China; Beijing Key laboratory for Quality Evaluation of Chinese Materia Medica, Beijing 100102, China
| | - Ruiqiang Liang
- National Institutes for Food and Drug Control, Beijing 100050, China
| | - Meilin Chen
- Beijing University of Chinese Medicine, Beijing 100102,China; Beijing Key laboratory for Quality Evaluation of Chinese Materia Medica, Beijing 100102, China
| | - Zhiqi Li
- Beijing University of Chinese Medicine, Beijing 100102,China; Beijing Key laboratory for Quality Evaluation of Chinese Materia Medica, Beijing 100102, China
| | - Xiaoyu Tao
- Beijing University of Chinese Medicine, Beijing 100102,China; Beijing Key laboratory for Quality Evaluation of Chinese Materia Medica, Beijing 100102, China
| | - Hongmin Ren
- Beijing University of Chinese Medicine, Beijing 100102,China; Beijing Key laboratory for Quality Evaluation of Chinese Materia Medica, Beijing 100102, China
| | - Yuhan Sheng
- Beijing University of Chinese Medicine, Beijing 100102,China
| | - Jiaqi Li
- Beijing University of Chinese Medicine, Beijing 100102,China; Beijing Key laboratory for Quality Evaluation of Chinese Materia Medica, Beijing 100102, China
| | - Ruichao Lin
- Beijing University of Chinese Medicine, Beijing 100102,China; Beijing Key laboratory for Quality Evaluation of Chinese Materia Medica, Beijing 100102, China.
| | - Chongjun Zhao
- Beijing University of Chinese Medicine, Beijing 100102,China; Beijing Key laboratory for Quality Evaluation of Chinese Materia Medica, Beijing 100102, China.
| | - Gaimei She
- Beijing University of Chinese Medicine, Beijing 100102,China.
| |
Collapse
|
10
|
Schöler D, Schnabl B. The role of the microbiome in liver disease. Curr Opin Gastroenterol 2024; 40:134-142. [PMID: 38362864 PMCID: PMC10990783 DOI: 10.1097/mog.0000000000001013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
PURPOSE OF REVIEW The intestinal microbiome and the gut-liver axis play a major role in health and disease. The human gut harbors trillions of microbes and a disruption of the gut homeostasis can contribute to liver disease. In this review, the progress in the field within the last 3 years is summarized, focusing on metabolic dysfunction-associated steatotic liver disease (MASLD), alcohol-associated liver disease (ALD), autoimmune liver disease (AILD), and hepatocellular carcinoma (HCC). RECENT FINDINGS Changes in the fecal virome and fungal mycobiome have been described in patients with various liver diseases. Several microbial derived metabolites including endogenous ethanol produced by bacteria, have been mechanistically linked to liver disease such as MASLD. Virulence factors encoded by gut bacteria contribute to ALD, AILD and HCC. Novel therapeutic approaches focused on the microbiome including phages, pre- and postbiotics have been successfully used in preclinical models. Fecal microbiota transplantation has been effective in attenuating liver disease. Probiotics are safe in patients with alcohol-associated hepatitis and improve liver disease and alcohol addiction. SUMMARY The gut-liver axis plays a key role in the pathophysiology of liver diseases. Understanding the microbiota in liver disease can help to develop precise microbiota centered therapies.
Collapse
Affiliation(s)
- David Schöler
- Department of Medicine, University of California, San Diego
| | - Bernd Schnabl
- Department of Medicine, University of California, San Diego
- Department of Medicine, VA San Diego Healthcare System, San Diego, California, USA
| |
Collapse
|
11
|
Stumme F, Steffens N, Steglich B, Mathies F, Nawrocki M, Sabihi M, Soukou-Wargalla S, Göke E, Kempski J, Fründt T, Weidemann S, Schramm C, Gagliani N, Huber S, Bedke T. A protective effect of inflammatory bowel disease on the severity of sclerosing cholangitis. Front Immunol 2024; 15:1307297. [PMID: 38510236 PMCID: PMC10950911 DOI: 10.3389/fimmu.2024.1307297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 02/12/2024] [Indexed: 03/22/2024] Open
Abstract
Background Primary sclerosing cholangitis (PSC) is a chronic liver disease marked by inflammation of the bile ducts and results in the development of strictures and fibrosis. A robust clinical correlation exists between PSC and inflammatory bowel disease (IBD). At present, published data are controversial, and it is yet unclear whether IBD drives or attenuates PSC. Methods Mdr2-deficient mice or DDC-fed mice were used as experimental models for sclerosing cholangitis. Additionally, colitis was induced in mice with experimental sclerosing cholangitis, either through infection with Citrobacter rodentium or by feeding with DSS. Lastly, fibrosis levels were determined through FibroScan analysis in people with PSC and PSC-IBD. Results Using two distinct experimental models of colitis and two models of sclerosing cholangitis, we found that colitis does not aggravate liver pathology, but rather reduces liver inflammation and liver fibrosis. Likewise, people with PSC-IBD have decreased liver fibrosis compared to those with PSC alone. Conclusions We found evidence that intestinal inflammation attenuates liver pathology. This study serves as a basis for further research on the pathogenesis of PSC and PSC-IBD, as well as the molecular mechanism responsible for the protective effect of IBD on PSC development. This study could lead to the discovery of novel therapeutic targets for PSC.
Collapse
Affiliation(s)
- Friederike Stumme
- Section of Molecular Immunology and Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Niklas Steffens
- Section of Molecular Immunology and Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Babett Steglich
- Section of Molecular Immunology and Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of General Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Franziska Mathies
- Section of Molecular Immunology and Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mikolaj Nawrocki
- Section of Molecular Immunology and Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Morsal Sabihi
- Section of Molecular Immunology and Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Shiwa Soukou-Wargalla
- Section of Molecular Immunology and Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Emilia Göke
- Section of Molecular Immunology and Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Kempski
- Section of Molecular Immunology and Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorben Fründt
- Section of Molecular Immunology and Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sören Weidemann
- Center of Diagnostics, Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph Schramm
- Section of Molecular Immunology and Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Martin Zeitz Center for Rare Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicola Gagliani
- Section of Molecular Immunology and Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of General Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Samuel Huber
- Section of Molecular Immunology and Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tanja Bedke
- Section of Molecular Immunology and Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
12
|
Trampert DC, Kunst RF, van de Graaf SFJ. Targeting bile salt homeostasis in biliary diseases. Curr Opin Gastroenterol 2024; 40:62-69. [PMID: 38230695 DOI: 10.1097/mog.0000000000000997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
PURPOSE OF REVIEW Advances in the understanding of bile salt synthesis, transport and signalling show the potential of modulating bile salt homeostasis as a therapeutic strategy in cholestatic liver diseases. Here, recent developments in (pre)clinical research in this field is summarized and discussed. RECENT FINDINGS Inhibition of the apical sodium-dependent bile salt transporter (ASBT) and Na + -taurocholate cotransporting polypeptide (NTCP) seems effective against cholestatic liver diseases, as well as Farnesoid X receptor (FXR) agonism or a combination of both. While approved for the treatment of primary biliary cholangitis (PBC) and intrahepatic cholestasis of pregnancy (ICP), ursodeoxycholic acid (UDCA) has retrospectively shown carefully promising results in primary sclerosing cholangitis (PSC). The side chain shortened derivate norUDCA is of further therapeutic interest since its mechanisms of action are independent of the bile salt transport machinery. In the pathogenesis of sclerosing cholangiopathies, a skewed T-cell response with alterations in gut microbiota and bile salt pool compositions are observed. In PSC pathogenesis, the bile salt receptor Takeda G-protein-coupled receptor 5 (TGR5) in cholangiocytes is implicated, whilst in immunoglobulin G4-related cholangitis the autoantigens annexin A11 and laminin 511-E8 are involved in protecting cholangiocytes. SUMMARY Modulating bile salt homeostasis has proven a promising treatment strategy in models of cholestasis and are continuously being further developed. Confirmatory clinical studies are needed in order to assess the proposed treatment strategies in patients allowing for a broader therapeutic arsenal in the future.
Collapse
Affiliation(s)
- David C Trampert
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, University of Amsterdam
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM), Amsterdam University Medical Centers
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Roni F Kunst
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, University of Amsterdam
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM), Amsterdam University Medical Centers
| | - Stan F J van de Graaf
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, University of Amsterdam
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM), Amsterdam University Medical Centers
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
13
|
Schneider CV, Alqahtani SA. Editorial: New Perspectives on Hepatic Steatosis. J Clin Exp Hepatol 2024; 14:101359. [PMID: 38406388 PMCID: PMC10884752 DOI: 10.1016/j.jceh.2024.101359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 02/01/2024] [Indexed: 02/27/2024] Open
Affiliation(s)
- Carolin V Schneider
- Department for Gastroenterology, Metabolic Disorders and Intensive Care Medicine (Medical Clinic III), RWTH Aachen University, Aachen, Germany
| | - Saleh A Alqahtani
- Organ Transplant Centre of Excellence, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
- Division of Gastroenterology & Hepatology, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
14
|
Özdirik B, Schnabl B. Microbial Players in Primary Sclerosing Cholangitis: Current Evidence and Concepts. Cell Mol Gastroenterol Hepatol 2023; 17:423-438. [PMID: 38109970 PMCID: PMC10837305 DOI: 10.1016/j.jcmgh.2023.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/12/2023] [Accepted: 12/12/2023] [Indexed: 12/20/2023]
Abstract
Primary sclerosing cholangitis (PSC) is a rare cholestatic liver disease with progressive biliary inflammation, destruction of the biliary tract, and fibrosis, resulting in liver cirrhosis and end-stage liver disease. To date, liver transplantation is the only definitive treatment option for PSC. The precise etiology of PSC remains elusive, but it is widely accepted to involve a complex interplay between genetic predisposition, immunologic dysfunction, and environmental influence. In recent years, the gut-liver axis has emerged as a crucial pathway contributing to the pathogenesis of PSC, with particular focus on the role of gut microbiota. However, the role of the fungal microbiome or mycobiome has been overlooked for years, resulting in a lack of comprehensive studies on its involvement in PSC. In this review, we clarify the present clinical and mechanistic data and concepts concerning the gut bacterial and fungal microbiota in the context of PSC. This review sheds light on the role of specific microbes and elucidates the dynamics of bacterial and fungal populations. Moreover, we discuss the latest insights into microbe-altering therapeutic approaches involving the gut-liver axis and bile acid metabolism.
Collapse
Affiliation(s)
- Burcin Özdirik
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, California; Department of Medicine, VA San Diego Healthcare System, San Diego, California.
| |
Collapse
|