1
|
Nagarajan V, Shi G, Horai R, Yu CR, Gopalakrishnan J, Yadav M, Liew MH, Gentilucci C, Caspi RR. IAN: An Intelligent System for Omics Data Analysis and Discovery. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.06.640921. [PMID: 40161796 PMCID: PMC11952324 DOI: 10.1101/2025.03.06.640921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
IAN is an R package that addresses the challenge of integrating, analyzing and interpreting high-throughput "omics" data, using a multi-agent artificial intelligence (AI) system. IAN leverages popular pathway and regulatory datasets (KEGG, WikiPathways, Reactome, GO, ChEA) and the STRING database for protein-protein interactions to perform standard enrichment analysis. The individual enrichment results are then used to generate insightful summaries, for each of the datasets, using a large language model (LLM) through a multi-agent architecture. These summaries are then contextually integrated and interpreted by the LLM, guided by carefully engineered prompts and grounding instructions, to provide insightful explanations, system overview, key regulators, novel observations etc. We demonstrate IAN's potential to facilitate biological discovery from complex omics data, by reanalyzing two already published data and evaluating the results. We also show remarkable performance of IAN, in terms of avoiding hallucination. IAN package, along with installation instructions and example usage, is available on https://github.com/NIH-NEI/IAN.
Collapse
Affiliation(s)
| | - Guangpu Shi
- Laboratory of Immunology, National Eye Institute, NIH, Bethesda 20892, USA
| | - Reiko Horai
- Laboratory of Immunology, National Eye Institute, NIH, Bethesda 20892, USA
| | - Cheng-Rong Yu
- Molecular Immunology Section, National Eye Institute, NIH, Bethesda 20892, USA
| | | | - Manoj Yadav
- Molecular Immunology Section, National Eye Institute, NIH, Bethesda 20892, USA
| | - Michael H Liew
- Neuro-Immune Regulome Unit, National Eye Institute, NIH, Bethesda 20892, USA
| | - Calla Gentilucci
- Laboratory of Immunology, National Eye Institute, NIH, Bethesda 20892, USA
| | - Rachel R Caspi
- Laboratory of Immunology, National Eye Institute, NIH, Bethesda 20892, USA
| |
Collapse
|
2
|
Croft M, Salek-Ardakani S, Ware CF. Targeting the TNF and TNFR superfamilies in autoimmune disease and cancer. Nat Rev Drug Discov 2024; 23:939-961. [PMID: 39448880 DOI: 10.1038/s41573-024-01053-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2024] [Indexed: 10/26/2024]
Abstract
The first anti-tumour necrosis factor (TNF) monoclonal antibody, infliximab (Remicade), celebrated its 25th anniversary of FDA approval in 2023. Inhibitors of TNF have since proved clinically efficacious at reducing inflammation associated with several autoimmune diseases, including rheumatoid arthritis, psoriasis and Crohn's disease. The success of TNF inhibitors raised unrealistic expectations for targeting other members of the TNF superfamily (TNFSF) of ligands and their receptors, with difficulties in part related to their more limited, variable expression and potential redundancy. However, there has been a resurgence of interest and investment, with many of these cytokines or their cognate receptors now under clinical investigation as targets for modulation of autoimmune and inflammatory diseases, as well as cancer. This Review assesses TNFSF-targeted biologics currently in clinical development for immune system-related diseases, highlighting ongoing challenges and future directions.
Collapse
Affiliation(s)
- Michael Croft
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, USA.
| | | | - Carl F Ware
- Laboratory of Molecular Immunology, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA.
| |
Collapse
|
3
|
Xu K, Kessler A, Nichetti F, Hoffmeister-Wittmann P, Scherr AL, Nader L, Kelmendi E, Schmitt N, Schwab M, García-Beccaria M, Sobol B, Nieto OA, Isele H, Gärtner U, Vaquero-Siguero N, Volk J, Korell F, Mock A, Heide D, Ramadori P, Lenoir B, Albrecht T, Hüllein J, Jäger D, Fröhling S, Springfeld C, Jackstadt R, Heikenwälder M, Dill MT, Roessler S, Goeppert B, Köhler BC. Lymphotoxin beta-activated LTBR/NIK/RELB axis drives proliferation in cholangiocarcinoma. Liver Int 2024; 44:2950-2963. [PMID: 39164890 DOI: 10.1111/liv.16069] [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: 03/21/2024] [Revised: 07/25/2024] [Accepted: 08/02/2024] [Indexed: 08/22/2024]
Abstract
BACKGROUND AND AIMS Cholangiocarcinoma (CCA) is an aggressive malignancy arising from the intrahepatic (iCCA) or extrahepatic (eCCA) bile ducts with poor prognosis and limited treatment options. Prior evidence highlighted a significant contribution of the non-canonical NF-κB signalling pathway in initiation and aggressiveness of different tumour types. Lymphotoxin-β (LTβ) stimulates the NF-κB-inducing kinase (NIK), resulting in the activation of the transcription factor RelB. However, the functional contribution of the non-canonical NF-κB signalling pathway via the LTβ/NIK/RelB axis in CCA carcinogenesis and progression has not been established. METHODS Human CCA-derived cell lines and organoids were examined to determine the expression of NF-κB pathway components upon activation or inhibition. Proliferation and cell death were analysed using real-time impedance measurement and flow cytometry. Immunoblot, qRT-PCR, RNA sequencing and in situ hybridization were employed to analyse gene and protein expression. Four in vivo models of iCCA were used to probe the activation and regulation of the non-canonical NF-κB pathway. RESULTS Exposure to LTα1/β2 activates the LTβ/NIK/RelB axis and promotes proliferation in CCA. Inhibition of NIK with the small molecule inhibitor B022 efficiently suppresses RelB expression in patient-derived CCA organoids and nuclear co-translocation of RelB and p52 stimulated by LTα1/β2 in CCA cell lines. In murine CCA, RelB expression is significantly increased and LTβ is the predominant ligand of the non-canonical NF-κB signalling pathway. CONCLUSIONS Our study confirms that the non-canonical NF-κB axis LTβ/NIK/RelB drives cholangiocarcinogenesis and represents a candidate therapeutic target.
Collapse
Affiliation(s)
- Kaiyu Xu
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
- Liver Cancer Center Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
- Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Annika Kessler
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
- Liver Cancer Center Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
- Department of Medicine A, Hematology, Oncology, Hemostaseology and Pneumology, University Hospital Münster, Münster, Germany
| | - Federico Nichetti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
- Computational Oncology, Molecular Diagnostics Program, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Paula Hoffmeister-Wittmann
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
- Liver Cancer Center Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
- Department of RadioOncology and Radiation Therapy, Heidelberg University Hospital, Heidelberg, Germany
| | - Anna-Lena Scherr
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
- Liver Cancer Center Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
| | - Luisa Nader
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
- Liver Cancer Center Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
| | - Eblina Kelmendi
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
- Liver Cancer Center Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
| | - Nathalie Schmitt
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
- Liver Cancer Center Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
| | - Maximilian Schwab
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
- Liver Cancer Center Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
| | - María García-Beccaria
- Division of Chronic Inflammation and Cancer, German Cancer Research Center Heidelberg (DKFZ), Heidelberg, Germany
- Madrid Institute for Advanced Study (MIAS), Madrid, Spain
| | - Benjamin Sobol
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
- Department of Gynecology and Obstetrics, University of Heidelberg, Heidelberg, Germany
| | - Osama Azzam Nieto
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
- Department of Gynecology and Obstetrics, University of Heidelberg, Heidelberg, Germany
| | - Hanna Isele
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
- Liver Cancer Center Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
- Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Ulrike Gärtner
- Interfaculty Biomedical Research Facility, University of Heidelberg, Heidelberg, Germany
| | - Nuria Vaquero-Siguero
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Cancer Progression and Metastasis Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Julia Volk
- Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Cancer Progression and Metastasis Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Felix Korell
- German Cancer Consortium (DKTK), Heidelberg, Germany
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Andreas Mock
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute for Pathology, Medical Faculty, Ludwig-Maximilians-University, Munich, Germany
| | - Danijela Heide
- Division of Chronic Inflammation and Cancer, German Cancer Research Center Heidelberg (DKFZ), Heidelberg, Germany
| | - Pierluigi Ramadori
- Division of Chronic Inflammation and Cancer, German Cancer Research Center Heidelberg (DKFZ), Heidelberg, Germany
| | - Bénédicte Lenoir
- Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center, Heidelberg, Germany
| | - Thomas Albrecht
- Liver Cancer Center Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
- Medical Faculty, Institute for Pathology, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
| | - Jennifer Hüllein
- Computational Oncology, Molecular Diagnostics Program, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
- Liver Cancer Center Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan Fröhling
- German Cancer Consortium (DKTK), Heidelberg, Germany
- Division of Translational Medical Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Christoph Springfeld
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
- Liver Cancer Center Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
| | - Rene Jackstadt
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Cancer Progression and Metastasis Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Mathias Heikenwälder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center Heidelberg (DKFZ), Heidelberg, Germany
- The M3 Research Center, Medical Faculty, University Clinic Tübingen (UKT), Tübingen, Germany
| | - Michael T Dill
- Liver Cancer Center Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
- Department of Gastroenterology, Infectious Diseases and Intoxication, Heidelberg University Hospital, Heidelberg, Germany
- Research Group Experimental Hepatology, Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stephanie Roessler
- Liver Cancer Center Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
- Medical Faculty, Institute for Pathology, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
| | - Benjamin Goeppert
- Institute of Pathology, RKH Klinikum Ludwigsburg, Ludwigsburg, Germany
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Bruno C Köhler
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
- Liver Cancer Center Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| |
Collapse
|
4
|
Cao H, Fu L, Liu D, Baranova A, Zhang F. Mendelian randomization analysis of causal and druggable circulating inflammatory proteins in schizophrenia. Front Psychiatry 2024; 15:1465291. [PMID: 39544374 PMCID: PMC11560794 DOI: 10.3389/fpsyt.2024.1465291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/09/2024] [Indexed: 11/17/2024] Open
Abstract
Background Schizophrenia (SZ) is a severe mental disorder with complex origins. Observational studies suggested that inflammatory factors may play a role in the pathophysiology of SZ and we aim to investigate the potential genetic connection between them by examining the causal impact of circulating inflammatory proteins on SZ. Methods We utilized Mendelian randomization (MR) analysis to assess the causal relationship between circulating inflammatory proteins and SZ and the GWAS summary datasets were sourced from public databases. The SZ dataset comprised 74,776 cases and 101,023 controls, while the summary results for 91 plasma proteins in 14,824 participants were obtained through the Olink Target platform. Moreover, to identify and evaluate potential drug targets, we searched the Drug-Gene Interaction Database (DGIdb). Results The results of the MR study confirmed that nine inflammatory proteins had a causal effect on SZ. Among these proteins, IL1A (OR: 0.93), TNFB (OR: 0.94), TNFSF14 (OR: 0.96), and CD40 (OR: 0.95) exhibited protective effects against SZ. Conversely, CCL23 (OR: 1.04), CCL19 (OR: 1.04), 4EBP1 (OR: 1.06), TWEAK (OR: 1.08), and DNER (OR: 1.10) were associated with an increased risk of SZ. The MR-Egger and weighted median methods also supported the direction of these effects. According to the Gene-Drug analysis, LTA, IL1A, CD40, and 4EBP1 can serve as drug targets. Conclusions Our study established causal relationships between circulating inflammatory proteins and SZ. It may be beneficial to personalize the treatment of SZ by incorporating inflammation management into the treatment regimen.
Collapse
Affiliation(s)
- Hongbao Cao
- School of Systems Biology, George Mason University, Manassas, VA, United States
| | - Li Fu
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dongming Liu
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Ancha Baranova
- School of Systems Biology, George Mason University, Manassas, VA, United States
- Research Centre for Medical Genetics, Moscow, Russia
| | - Fuquan Zhang
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Institute of Neuropsychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
5
|
Chen Y, Yan Y, Li Y, Zhang L, Luo T, Zhu X, Qin D, Chen N, Huang W, Chen X, Wang L, Zhu X, Zhang L. Deletion of Tgm2 suppresses BMP-mediated hepatocyte-to-cholangiocyte metaplasia in ductular reaction. Cell Prolif 2024; 57:e13646. [PMID: 38623945 PMCID: PMC11471396 DOI: 10.1111/cpr.13646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/26/2024] [Accepted: 04/03/2024] [Indexed: 04/17/2024] Open
Abstract
Transglutaminase 2 (Tgm2) plays an essential role in hepatic repair following prolonged toxic injury. During cholestatic liver injury, the intrahepatic cholangiocytes undergo dynamic tissue expansion and remodelling, referred to as ductular reaction (DR), which is crucial for liver regeneration. However, the molecular mechanisms governing the dynamics of active cells in DR are still largely unclear. Here, we generated Tgm2-knockout mice (Tgm2-/-) and Tgm2-CreERT2-Rosa26-mTmG flox/flox (Tgm2CreERT2-R26T/Gf/f) mice and performed a three-dimensional (3D) collagen gel culture of mouse hepatocytes to demonstrate how Tgm2 signalling is involved in DR to remodel intrahepatic cholangiocytes. Our results showed that the deletion of Tgm2 adversely affected the functionality and maturity of the proliferative cholangiocytes in DR, thus leading to more severe cholestasis during DDC-induced liver injury. Additionally, Tgm2 hepatocytes played a crucial role in the regulation of DR through metaplasia. We unveiled that Tgm2 regulated H3K4me3Q5ser via serotonin to promote BMP signalling activation to participate in DR. Besides, we revealed that the activation or inhibition of BMP signalling could promote or suppress the development and maturation of cholangiocytes in DDC-induced DR. Furthermore, our 3D collagen gel culture assay indicated that Tgm2 was vital for the development of cholangiocytes in vitro. Our results uncovered a considerable role of BMP signalling in controlling metaplasia of Tgm2 hepatocytes in DR and revealed the phenotypic plasticity of mature hepatocytes.
Collapse
Affiliation(s)
- Yaqing Chen
- College of Veterinary Medicine/College of Biomedicine and HealthHuazhong Agricultural UniversityWuhanChina
| | - Yi Yan
- College of Veterinary Medicine/College of Biomedicine and HealthHuazhong Agricultural UniversityWuhanChina
| | - Yujing Li
- College of Veterinary Medicine/College of Biomedicine and HealthHuazhong Agricultural UniversityWuhanChina
| | - Liang Zhang
- College of Veterinary Medicine/College of Biomedicine and HealthHuazhong Agricultural UniversityWuhanChina
| | - Tingting Luo
- College of Veterinary Medicine/College of Biomedicine and HealthHuazhong Agricultural UniversityWuhanChina
| | - Xinlong Zhu
- College of Veterinary Medicine/College of Biomedicine and HealthHuazhong Agricultural UniversityWuhanChina
| | - Dan Qin
- College of Veterinary Medicine/College of Biomedicine and HealthHuazhong Agricultural UniversityWuhanChina
| | - Ning Chen
- College of Veterinary Medicine/College of Biomedicine and HealthHuazhong Agricultural UniversityWuhanChina
| | - Wendong Huang
- Department of Diabetes Complications and MetabolismDiabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical CenterDuarteCaliforniaUSA
| | - Xiangmei Chen
- Department of Nephrology, First Medical Center of Chinese PLA General HospitalNephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease ResearchBeijingChina
| | - Liqiang Wang
- Department of Nephrology, First Medical Center of Chinese PLA General HospitalNephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease ResearchBeijingChina
| | - Xianmin Zhu
- Department of Hepatobiliary and Pancreatic SurgeryCancer Hospital of Wuhan University (Hubei Cancer Hospital)WuhanChina
| | - Lisheng Zhang
- College of Veterinary Medicine/College of Biomedicine and HealthHuazhong Agricultural UniversityWuhanChina
| |
Collapse
|
6
|
Jalan-Sakrikar N, Guicciardi ME, O’Hara SP, Azad A, LaRusso NF, Gores GJ, Huebert RC. Central role for cholangiocyte pathobiology in cholestatic liver diseases. Hepatology 2024:01515467-990000000-01022. [PMID: 39250501 PMCID: PMC11890218 DOI: 10.1097/hep.0000000000001093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 08/21/2024] [Indexed: 09/11/2024]
Abstract
Cholangiopathies comprise a spectrum of chronic intrahepatic and extrahepatic biliary tract disorders culminating in progressive cholestatic liver injury, fibrosis, and often cirrhosis and its sequela. Treatment for these diseases is limited, and collectively, they are one of the therapeutic "black boxes" in clinical hepatology. The etiopathogenesis of the cholangiopathies likely includes disease-specific mediators but also common cellular and molecular events driving disease progression (eg, cholestatic fibrogenesis, inflammation, and duct damage). The common pathways involve cholangiocytes, the epithelial cells lining the intrahepatic and extrahepatic bile ducts, which are central to the pathogenesis of these disorders. Current information suggests that cholangiocytes function as a signaling "hub" in biliary tract-associated injury. Herein, we review the pivotal role of cholangiocytes in cholestatic fibrogenesis, focusing on the crosstalk between cholangiocytes and portal fibroblasts and HSCs. The proclivity of these cells to undergo a senescence-associated secretory phenotype, which is proinflammatory and profibrogenic, and the intrinsic intracellular activation pathways resulting in the secretion of cytokines and chemokines are reviewed. The crosstalk between cholangiocytes and cells of the innate (neutrophils and macrophages) and adaptive (T cells and B cells) immune systems is also examined in detail. The information will help consolidate information on this topic and guide further research and potential therapeutic strategies for these diseases.
Collapse
Affiliation(s)
- Nidhi Jalan-Sakrikar
- Division of Gastroenterology and Hepatology, Mayo College of Medicine and Science, Mayo Clinic, Rochester, MN
- Gastroenterology Research Unit, Mayo College of Medicine and Science, Mayo Clinic, Rochester, MN
- Mayo Clinic Center for Cell Signaling in Gastroenterology; Mayo College of Medicine and Science, Mayo Clinic, Rochester, MN
| | - Maria Eugenia Guicciardi
- Division of Gastroenterology and Hepatology, Mayo College of Medicine and Science, Mayo Clinic, Rochester, MN
| | - Steven P. O’Hara
- Division of Gastroenterology and Hepatology, Mayo College of Medicine and Science, Mayo Clinic, Rochester, MN
| | - Adiba Azad
- Division of Gastroenterology and Hepatology, Mayo College of Medicine and Science, Mayo Clinic, Rochester, MN
- Mayo Clinic Center for Cell Signaling in Gastroenterology; Mayo College of Medicine and Science, Mayo Clinic, Rochester, MN
| | - Nicholas F. LaRusso
- Division of Gastroenterology and Hepatology, Mayo College of Medicine and Science, Mayo Clinic, Rochester, MN
- Mayo Clinic Center for Cell Signaling in Gastroenterology; Mayo College of Medicine and Science, Mayo Clinic, Rochester, MN
| | - Gregory J. Gores
- Division of Gastroenterology and Hepatology, Mayo College of Medicine and Science, Mayo Clinic, Rochester, MN
- Mayo Clinic Center for Cell Signaling in Gastroenterology; Mayo College of Medicine and Science, Mayo Clinic, Rochester, MN
| | - Robert C. Huebert
- Division of Gastroenterology and Hepatology, Mayo College of Medicine and Science, Mayo Clinic, Rochester, MN
- Gastroenterology Research Unit, Mayo College of Medicine and Science, Mayo Clinic, Rochester, MN
- Mayo Clinic Center for Cell Signaling in Gastroenterology; Mayo College of Medicine and Science, Mayo Clinic, Rochester, MN
| |
Collapse
|
7
|
Scherr AL, Nader L, Xu K, Elssner C, Ridder DA, Nichetti F, Mastel M, Fritzsche S, Kelmendi E, Schmitt N, Hoffmeister-Wittmann P, Weiler SME, Korell F, Albrecht T, Schwab M, Isele H, Kessler A, Hüllein J, Seretny A, Ye L, Urbanik T, Welte S, Leblond AL, Heilig CE, Rahbari M, Ali A, Gallage S, Lenoir B, Wilhelm N, Gärtner U, Ogrodnik SJ, Springfeld C, Tschaharganeh D, Fröhling S, Longerich T, Schulze-Bergkamen H, Jäger D, Brandl L, Schirmacher P, Straub BK, Weber A, De Toni EN, Goeppert B, Heikenwalder M, Jackstadt R, Roessler S, Breuhahn K, Köhler BC. Etiology-independent activation of the LTβ-LTβR-RELB axis drives aggressiveness and predicts poor prognosis in HCC. Hepatology 2024; 80:278-294. [PMID: 37916976 DOI: 10.1097/hep.0000000000000657] [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] [Received: 12/21/2021] [Accepted: 09/21/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND AND AIMS HCC is the most common primary liver tumor, with an increasing incidence worldwide. HCC is a heterogeneous malignancy and usually develops in a chronically injured liver. The NF-κB signaling network consists of a canonical and a noncanonical branch. Activation of canonical NF-κB in HCC is documented. However, a functional and clinically relevant role of noncanonical NF-κB and its downstream effectors is not established. APPROACH AND RESULTS Four human HCC cohorts (total n = 1462) and 4 mouse HCC models were assessed for expression and localization of NF-κB signaling components and activating ligands. In vitro , NF-κB signaling, proliferation, and cell death were measured, proving a pro-proliferative role of v-rel avian reticuloendotheliosis viral oncogene homolog B (RELB) activated by means of NF-κB-inducing kinase. In vivo , lymphotoxin beta was identified as the predominant inducer of RELB activation. Importantly, hepatocyte-specific RELB knockout in a murine HCC model led to a lower incidence compared to controls and lower maximal tumor diameters. In silico , RELB activity and RELB-directed transcriptomics were validated on the The Cancer Genome Atlas HCC cohort using inferred protein activity and Gene Set Enrichment Analysis. In RELB-active HCC, pathways mediating proliferation were significantly activated. In contrast to v-rel avian reticuloendotheliosis viral oncogene homolog A, nuclear enrichment of noncanonical RELB expression identified patients with a poor prognosis in an etiology-independent manner. Moreover, RELB activation was associated with malignant features metastasis and recurrence. CONCLUSIONS This study demonstrates a prognostically relevant, etiology-independent, and cross-species consistent activation of a lymphotoxin beta/LTβR/RELB axis in hepatocarcinogenesis. These observations may harbor broad implications for HCC, including possible clinical exploitation.
Collapse
Affiliation(s)
- Anna-Lena Scherr
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Luisa Nader
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Kaiyu Xu
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Christin Elssner
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Dirk A Ridder
- Department of General Pathology, University Hospital Mainz, Mainz, Germany
| | - Federico Nichetti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
- Computational Oncology, Molecular Diagnostics Program, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Manuel Mastel
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Cancer Progression and Metastasis Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Sarah Fritzsche
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Eblina Kelmendi
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Nathalie Schmitt
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Paula Hoffmeister-Wittmann
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
- Department of Radiooncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Sofia M E Weiler
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Felix Korell
- German Cancer Consortium (DKTK), Heidelberg, Germany
- Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Thomas Albrecht
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Liver Cancer Center Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Maximilian Schwab
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Hanna Isele
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Annika Kessler
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Jennifer Hüllein
- Computational Oncology, Molecular Diagnostics Program, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Agnieszka Seretny
- Helmholtz-University Group "Cell Plasticity and Epigenetic Remodeling", German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Liangtao Ye
- Department of Internal Medicine II, Klinikum Grosshadern, Ludwig-Maximilians-University of Munich
| | - Toni Urbanik
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Stefan Welte
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
- Department of Radiooncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Anne-Laure Leblond
- Department of Pathology and Molecular Pathology, University and University Hospital Zurich, Zurich, Switzerland
| | - Christoph E Heilig
- German Cancer Consortium (DKTK), Heidelberg, Germany
- Department of Translational Medical Oncology, NCT Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mohammad Rahbari
- Division of Chronic Inflammation and Cancer, German Cancer Research Center Heidelberg (DKFZ), Heidelberg, Germany
| | - Adnan Ali
- Division of Chronic Inflammation and Cancer, German Cancer Research Center Heidelberg (DKFZ), Heidelberg, Germany
| | - Suchira Gallage
- Division of Chronic Inflammation and Cancer, German Cancer Research Center Heidelberg (DKFZ), Heidelberg, Germany
| | - Bénédicte Lenoir
- Clinical Cooperation Unit "Applied Tumor Immunity", German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Nina Wilhelm
- Clinical Cooperation Unit "Applied Tumor Immunity", German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Ulrike Gärtner
- Interfaculty Biomedical Research Facility, University of Heidelberg, Heidelberg, Germany
| | - Simon J Ogrodnik
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christoph Springfeld
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
- Liver Cancer Center Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Darjus Tschaharganeh
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Helmholtz-University Group "Cell Plasticity and Epigenetic Remodeling", German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan Fröhling
- German Cancer Consortium (DKTK), Heidelberg, Germany
- Department of Translational Medical Oncology, NCT Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thomas Longerich
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Lydia Brandl
- Institute of Pathology, Medical Faculty, Ludwig-Maximilians-University, Munich, Germany
| | - Peter Schirmacher
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Liver Cancer Center Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Beate K Straub
- Department of General Pathology, University Hospital Mainz, Mainz, Germany
| | - Achim Weber
- Department of Pathology and Molecular Pathology, University and University Hospital Zurich, Zurich, Switzerland
| | - Enrico N De Toni
- Department of Internal Medicine II, Klinikum Grosshadern, Ludwig-Maximilians-University of Munich
| | - Benjamin Goeppert
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Liver Cancer Center Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center Heidelberg (DKFZ), Heidelberg, Germany
- The M3 Research Center, University Clinic Tübingen (UKT), Medical faculty, Tübingen, Germany
| | - Rene Jackstadt
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Cancer Progression and Metastasis Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Stephanie Roessler
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Liver Cancer Center Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Kai Breuhahn
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Liver Cancer Center Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Bruno C Köhler
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- Liver Cancer Center Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
8
|
Zhang J, Lyu Z, Li B, You Z, Cui N, Li Y, Li Y, Huang B, Chen R, Chen Y, Peng Y, Fang J, Wang Q, Miao Q, Tang R, Gershwin ME, Lian M, Xiao X, Ma X. P4HA2 induces hepatic ductular reaction and biliary fibrosis in chronic cholestatic liver diseases. Hepatology 2023; 78:10-25. [PMID: 36799463 DOI: 10.1097/hep.0000000000000317] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 01/06/2023] [Indexed: 02/18/2023]
Abstract
BACKGROUNDS Prolyl-4-hydroxylases (P4Hs) are key enzymes in collagen synthesis. The P4HA subunit (P4HA1, P4HA2, and P4HA3) contains a substrate binding and catalyzation domain. We postulated that P4HA2 would play a key role in the cholangiocyte pathology of cholestatic liver diseases. METHODS We studied humans with primary biliary cholangitis (PBC) and Primary sclerosing cholangitis (PSC), P4HA2 -/- mice injured by DDC, and P4HA2 -/- /MDR2 -/- double knockout mice. A parallel study was performed in patients with PBC, PSC, and controls using immunohistochemistry and immunofluorescence. In the murine model, the level of ductular reaction and biliary fibrosis were monitored by histology, qPCR, immunohistochemistry, and Western blotting. Expression of Yes1 Associated Transcriptional Regulator (YAP) phosphorylation was measured in isolated mouse cholangiocytes. The mechanism of P4HA2 was explored in RBE and 293T cell lines by using qPCR, Western blot, immunofluorescence, and co-immunoprecipitation. RESULTS The hepatic expression level of P4HA2 was highly elevated in patients with PBC or PSC. Ductular reactive cholangiocytes predominantly expressed P4HA2. Cholestatic patients with more severe liver injury correlated with levels of P4HA2 in the liver. In P4HA2 -/- mice, there was a significantly reduced level of ductular reaction and fibrosis compared with controls in the DDC-induced chronic cholestasis. Decreased liver fibrosis and ductular reaction were observed in P4HA2 -/- /MDR2 -/- mice compared with MDR2 -/- mice. Cholangiocytes isolated from P4HA2 -/- /MDR2 -/- mice displayed a higher level of YAP phosphorylation, resulting in cholangiocytes proliferation inhibition. In vitro studies showed that P4HA2 promotes RBE cell proliferation by inducing SAV1 degradation, eventually resulting in the activation of YAP. CONCLUSIONS P4HA2 promotes hepatic ductular reaction and biliary fibrosis by regulating the SAV1-mediated Hippo signaling pathway. P4HA2 is a potential therapeutic target for PBC and PSC.
Collapse
Affiliation(s)
- Jun Zhang
- Division of Gastroenterology and Hepatology, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai Institute of Digestive Disease, Middle Shandong Road, Shanghai 200001, China
| | - Zhuwan Lyu
- Division of Gastroenterology and Hepatology, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai Institute of Digestive Disease, Middle Shandong Road, Shanghai 200001, China
| | - Bo Li
- Division of Gastroenterology and Hepatology, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai Institute of Digestive Disease, Middle Shandong Road, Shanghai 200001, China
| | - Zhengrui You
- Division of Gastroenterology and Hepatology, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai Institute of Digestive Disease, Middle Shandong Road, Shanghai 200001, China
| | - Nana Cui
- Division of Gastroenterology and Hepatology, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai Institute of Digestive Disease, Middle Shandong Road, Shanghai 200001, China
| | - You Li
- Division of Gastroenterology and Hepatology, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai Institute of Digestive Disease, Middle Shandong Road, Shanghai 200001, China
| | - Yikang Li
- Division of Gastroenterology and Hepatology, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai Institute of Digestive Disease, Middle Shandong Road, Shanghai 200001, China
| | - Bingyuan Huang
- Division of Gastroenterology and Hepatology, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai Institute of Digestive Disease, Middle Shandong Road, Shanghai 200001, China
| | - Ruiling Chen
- Division of Gastroenterology and Hepatology, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai Institute of Digestive Disease, Middle Shandong Road, Shanghai 200001, China
| | - Yong Chen
- Division of Gastroenterology and Hepatology, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai Institute of Digestive Disease, Middle Shandong Road, Shanghai 200001, China
| | - Yanshen Peng
- Division of Gastroenterology and Hepatology, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai Institute of Digestive Disease, Middle Shandong Road, Shanghai 200001, China
| | - Jingyuan Fang
- Division of Gastroenterology and Hepatology, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai Institute of Digestive Disease, Middle Shandong Road, Shanghai 200001, China
| | - Qixia Wang
- Division of Gastroenterology and Hepatology, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai Institute of Digestive Disease, Middle Shandong Road, Shanghai 200001, China
| | - Qi Miao
- Division of Gastroenterology and Hepatology, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai Institute of Digestive Disease, Middle Shandong Road, Shanghai 200001, China
| | - Ruqi Tang
- Division of Gastroenterology and Hepatology, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai Institute of Digestive Disease, Middle Shandong Road, Shanghai 200001, China
| | - M Eric Gershwin
- Division of Rheumatology, Department of Medicine, Allergy and Clinical Immunology, University of California at Davis, Davis, California, USA
| | - Min Lian
- Division of Gastroenterology and Hepatology, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai Institute of Digestive Disease, Middle Shandong Road, Shanghai 200001, China
| | - Xiao Xiao
- Division of Gastroenterology and Hepatology, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai Institute of Digestive Disease, Middle Shandong Road, Shanghai 200001, China
| | - Xiong Ma
- Division of Gastroenterology and Hepatology, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai Institute of Digestive Disease, Middle Shandong Road, Shanghai 200001, China
| |
Collapse
|
9
|
Hoffmeister-Wittmann P, Mock A, Nichetti F, Korell F, Heilig CE, Scherr AL, Günther M, Albrecht T, Kelmendi E, Xu K, Nader L, Kessler A, Schmitt N, Fritzsche S, Weiler S, Sobol B, Stenzinger A, Boeck S, Westphalen CB, Schulze-Osthoff K, Trojan J, Kindler T, Weichert W, Spiekermann K, Bitzer M, Folprecht G, Illert AL, Boerries M, Klauschen F, Ochsenreither S, Siveke J, Bauer S, Glimm H, Brors B, Hüllein J, Hübschmann D, Uhrig S, Horak P, Kreutzfeldt S, Banales JM, Springfeld C, Jäger D, Schirmacher P, Roessler S, Ormanns S, Goeppert B, Fröhling S, Köhler BC. Bcl-x L as prognostic marker and potential therapeutic target in cholangiocarcinoma. Liver Int 2022; 42:2855-2870. [PMID: 35983950 DOI: 10.1111/liv.15392] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 07/26/2022] [Accepted: 08/08/2022] [Indexed: 01/27/2023]
Abstract
Intrahepatic, perihilar, and distal cholangiocarcinoma (iCCA, pCCA, dCCA) are highly malignant tumours with increasing mortality rates due to therapy resistances. Among the mechanisms mediating resistance, overexpression of anti-apoptotic Bcl-2 proteins (Bcl-2, Bcl-xL , Mcl-1) is particularly important. In this study, we investigated whether antiapoptotic protein patterns are prognostically relevant and potential therapeutic targets in CCA. Bcl-2 proteins were analysed in a pan-cancer cohort from the NCT/DKFZ/DKTK MASTER registry trial (n = 1140, CCA n = 72) via RNA-sequencing and transcriptome-based protein activity interference revealing high ranks of CCA for Bcl-xL and Mcl-1. Expression of Bcl-xL , Mcl-1, and Bcl-2 was assessed in human CCA tissue and cell lines compared with cholangiocytes by immunohistochemistry, immunoblotting, and quantitative-RT-PCR. Immunohistochemistry confirmed the upregulation of Bcl-xL and Mcl-1 in iCCA tissues. Cell death of CCA cell lines upon treatment with specific small molecule inhibitors of Bcl-xL (Wehi-539), of Mcl-1 (S63845), and Bcl-2 (ABT-199), either alone, in combination with each other or together with chemotherapeutics was assessed by flow cytometry. Targeting Bcl-xL induced cell death and augmented the effect of chemotherapy in CCA cells. Combined inhibition of Bcl-xL and Mcl-1 led to a synergistic increase in cell death in CCA cell lines. Correlation between Bcl-2 protein expression and survival was analysed within three independent patient cohorts from cancer centers in Germany comprising 656 CCA cases indicating a prognostic value of Bcl-xL in CCA depending on the CCA subtype. Collectively, these observations identify Bcl-xL as a key protein in cell death resistance of CCA and may pave the way for clinical application.
Collapse
Affiliation(s)
- Paula Hoffmeister-Wittmann
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, University Hospital Heidelberg, Heidelberg, Germany.,Department of Radiooncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Andreas Mock
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, University Hospital Heidelberg, Heidelberg, Germany.,Department of Translational Medical Oncology, NCT Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg and Partner Sites, Heidelberg, Germany.,Institute of Pathology, Medical Faculty, Ludwig-Maximilians-University, Munich, Germany
| | - Federico Nichetti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy.,Computational Oncology Group, Molecular Precision Oncology Program, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Felix Korell
- German Cancer Consortium (DKTK), Heidelberg and Partner Sites, Heidelberg, Germany.,Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Christoph E Heilig
- Department of Translational Medical Oncology, NCT Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg and Partner Sites, Heidelberg, Germany
| | - Anna-Lena Scherr
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Michael Günther
- German Cancer Consortium (DKTK), Heidelberg and Partner Sites, Heidelberg, Germany.,Institute of Pathology, Medical Faculty, Ludwig-Maximilians-University, Munich, Germany
| | - Thomas Albrecht
- Institute for Pathology, University Hospital Heidelberg, Heidelberg, Germany.,Liver Cancer Center Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Eblina Kelmendi
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Kaiyu Xu
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Luisa Nader
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Annika Kessler
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Nathalie Schmitt
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Sarah Fritzsche
- Institute for Pathology, University Hospital Heidelberg, Heidelberg, Germany.,Liver Cancer Center Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Sofia Weiler
- Institute for Pathology, University Hospital Heidelberg, Heidelberg, Germany.,Liver Cancer Center Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Benjamin Sobol
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Albrecht Stenzinger
- Institute for Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Stefan Boeck
- German Cancer Consortium (DKTK), Heidelberg and Partner Sites, Heidelberg, Germany.,Department of Internal Medicine III and Comprehensive Cancer Center, Klinikum Grosshadern, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Christoph B Westphalen
- German Cancer Consortium (DKTK), Heidelberg and Partner Sites, Heidelberg, Germany.,Department of Internal Medicine III and Comprehensive Cancer Center, Klinikum Grosshadern, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Klaus Schulze-Osthoff
- German Cancer Consortium (DKTK), Heidelberg and Partner Sites, Heidelberg, Germany.,Department of Molecular Medicine, Interfaculty Institute for Biochemistry, University of Tübingen, Tübingen, Germany
| | - Jörg Trojan
- German Cancer Consortium (DKTK), Heidelberg and Partner Sites, Heidelberg, Germany.,Department of Gastroenterology, Gastrointestinal Medical Oncology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Thomas Kindler
- German Cancer Consortium (DKTK), Heidelberg and Partner Sites, Heidelberg, Germany.,University Cancer Center, University Medical Center Mainz, Germany
| | - Wilko Weichert
- German Cancer Consortium (DKTK), Heidelberg and Partner Sites, Heidelberg, Germany.,Institute of Pathology, Medical Faculty, Technichal University Munich, Munich, Germany
| | - Karsten Spiekermann
- Department of Translational Medical Oncology, NCT Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg and Partner Sites, Heidelberg, Germany.,Department of Hematology and Medical Oncology, University Hospital Munich, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Michael Bitzer
- German Cancer Consortium (DKTK), Heidelberg and Partner Sites, Heidelberg, Germany.,Department of Internal Medicine I, University Hospital Tübingen, Tübingen, Germany
| | - Gunnar Folprecht
- German Cancer Consortium (DKTK), Heidelberg and Partner Sites, Heidelberg, Germany.,Department of Hematology and Medical Oncology, Carl Gustav Carus University Hospital, Dresden, Germany
| | - Anna L Illert
- German Cancer Consortium (DKTK), Heidelberg and Partner Sites, Heidelberg, Germany.,Department of Internal Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Comprehensive Cancer Center Freiburg (CCCF), Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Melanie Boerries
- German Cancer Consortium (DKTK), Heidelberg and Partner Sites, Heidelberg, Germany.,Comprehensive Cancer Center Freiburg (CCCF), Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Frederick Klauschen
- German Cancer Consortium (DKTK), Heidelberg and Partner Sites, Heidelberg, Germany.,Institute of Pathology, Charité University Medicine Berlin, Berlin, Germany
| | - Sebastian Ochsenreither
- German Cancer Consortium (DKTK), Heidelberg and Partner Sites, Heidelberg, Germany.,Faculty of Medicine, Charité Comprehensive Cancer Center (CCCC), Humboldt University of Berlin, Berlin, Germany.,Department of hematology, medical oncology and tumor immunology, Charité University Medicine Berlin, Berlin, Germany
| | - Jens Siveke
- German Cancer Consortium (DKTK), Heidelberg and Partner Sites, Heidelberg, Germany.,Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK Partner Site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - Sebastian Bauer
- German Cancer Consortium (DKTK), Heidelberg and Partner Sites, Heidelberg, Germany.,Department for Translational Oncology, West German Tumor Center (WTZ), Essen University Hospital, Essen, Germany
| | - Hanno Glimm
- Translational Medical Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Department for Translational Medical Oncology, National Center for Tumor Diseases (NCT/UCC), Dresden, Germany: Germany Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany.,German Cancer Consortium (DKTK) Dresden, Germany.,Translational Functional Cancer Genomics, National Center für Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Benedikt Brors
- Computational Oncology Group, Molecular Precision Oncology Program, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jennifer Hüllein
- Computational Oncology Group, Molecular Precision Oncology Program, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniel Hübschmann
- German Cancer Consortium (DKTK), Heidelberg and Partner Sites, Heidelberg, Germany.,Computational Oncology Group, Molecular Precision Oncology Program, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Heidelberg Institute for Stem cell Technology and Experimental Medicine (HI-STEM), Heidelberg, Germany
| | - Sebastian Uhrig
- Computational Oncology Group, Molecular Precision Oncology Program, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Horak
- Department of Translational Medical Oncology, NCT Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg and Partner Sites, Heidelberg, Germany
| | - Simon Kreutzfeldt
- Department of Translational Medical Oncology, NCT Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg and Partner Sites, Heidelberg, Germany
| | - Jesus M Banales
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute, Ikerbasque, Network Centre for Biomedical Research in Liver and Digestive Diseases (CIBERehd), San Sebastian, Spain.,Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Christoph Springfeld
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Peter Schirmacher
- Institute for Pathology, University Hospital Heidelberg, Heidelberg, Germany.,Liver Cancer Center Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Stephanie Roessler
- Institute for Pathology, University Hospital Heidelberg, Heidelberg, Germany.,Liver Cancer Center Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Steffen Ormanns
- German Cancer Consortium (DKTK), Heidelberg and Partner Sites, Heidelberg, Germany.,Institute of Pathology, Medical Faculty, Ludwig-Maximilians-University, Munich, Germany
| | - Benjamin Goeppert
- Institute for Pathology, University Hospital Heidelberg, Heidelberg, Germany.,Liver Cancer Center Heidelberg, University Hospital Heidelberg, Heidelberg, Germany.,Institute of Pathology and Neuropathology, RKH Klinikum Ludwigsburg, Ludwigsburg, Germany
| | - Stefan Fröhling
- Department of Translational Medical Oncology, NCT Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg and Partner Sites, Heidelberg, Germany
| | - Bruno C Köhler
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, University Hospital Heidelberg, Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg and Partner Sites, Heidelberg, Germany.,Liver Cancer Center Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
10
|
Yuan Z, Wang J, Zhang H, Miao Y, Tang Q, Yuan Z, Nong C, Duan Z, Zhang L, Jiang Z, Yu Q. Triptolide increases resistance to bile duct ligation-induced liver injury and fibrosis in mice by inhibiting RELB. Front Nutr 2022; 9:1032722. [PMID: 36313114 PMCID: PMC9608656 DOI: 10.3389/fnut.2022.1032722] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 09/28/2022] [Indexed: 12/04/2022] Open
Abstract
Cholestasis is a common, chronic liver disease that may cause fibrosis and cirrhosis. Tripterygium wilfordii Hook.f (TWHF) is a species in the Euonymus family that is commonly used as a source of medicine and food in Eastern and Southern China. Triptolide (TP) is an epoxy diterpene lactone of TWHF, as well as the main active ingredient in TWHF. Here, we used a mouse model of common bile duct ligation (BDL) cholestasis, along with cultured human intrahepatic biliary epithelial cells, to explore whether TP can relieve cholestasis. Compared with the control treatment, TP at a dose of 70 or 140 μg/kg reduced the serum levels of the liver enzymes alanine transaminase, aspartate aminotransferase, and alkaline phosphatase in mice; hematoxylin and eosin staining also showed that TP reduced necrosis in tissues. Both in vitro and in vivo analyses revealed that TP inhibited cholangiocyte proliferation by reducing the expression of RelB. Immunohistochemical staining of CK19 and Ki67, as well as measurement of Ck19 mRNA levels in hepatic tissue, revealed that TP inhibited the BDL-induced ductular reaction. Masson 3 and Sirius Red staining for hepatic hydroxyproline showed that TP alleviated BDL-induced hepatic fibrosis. Additionally, TP substantially inhibited BDL-induced hepatic inflammation. In summary, TP inhibited the BDL-induced ductular reaction by reducing the expression of RelB in cholangiocytes, thereby alleviating liver injury, fibrosis, and inflammation.
Collapse
Affiliation(s)
- Zihang Yuan
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jie Wang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Haoran Zhang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Yingying Miao
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Qianhui Tang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Ziqiao Yuan
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Cheng Nong
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Zhicheng Duan
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Luyong Zhang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China,Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhenzhou Jiang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China,Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, China,*Correspondence: Zhenzhou Jiang,
| | - Qinwei Yu
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China,Qinwei Yu,
| |
Collapse
|
11
|
Biliary NIK promotes ductular reaction and liver injury and fibrosis in mice. Nat Commun 2022; 13:5111. [PMID: 36042192 PMCID: PMC9427946 DOI: 10.1038/s41467-022-32575-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 08/08/2022] [Indexed: 11/21/2022] Open
Abstract
Excessive cholangiocyte expansion (ductular reaction) promotes liver disease progression, but the underlying mechanism is poorly understood. Here we identify biliary NF-κB-inducing kinase (NIK) as a pivotal regulator of ductular reaction. NIK is known to activate the noncanonical IKKα/NF-κB2 pathway and regulate lymphoid tissue development. We find that cholangiocyte NIK is upregulated in mice with cholestasis induced by bile duct ligation (BDL), 5-diethoxycarbonyl-1,4-dihydrocollidine (DDC), or α-naphtyl-isothiocyanate (ANIT). DDC, ANIT, or BDL induces ductular reaction, liver injury, inflammation, and fibrosis in mice. Cholangiocyte-specific deletion of NIK, but not IKKα, blunts these pathological alterations. NIK inhibitor treatment similarly ameliorates DDC-induced ductular reaction, liver injury, and fibrosis. Biliary NIK directly increases cholangiocyte proliferation while suppressing cholangiocyte death, and it also promotes secretion of cholangiokines from cholangiocytes. Cholangiokines stimulate liver macrophages and hepatic stellate cells, augmenting liver inflammation and fibrosis. These results unveil a NIK/ductular reaction axis and a NIK/cholangiokine axis that promote liver disease progression. Excessive expansion of cholangiocytes in the liver leads to ductular reaction and liver disease. Here, the authors show that genetic ablation, or pharmacological inhibition, of biliary NIK blocks ductular reaction, liver inflammation, and liver fibrosis in mice by modulating secretion of cholangiokines that mediate liver inflammation and fibrosis.
Collapse
|
12
|
Tao L, Ren X, Zhai W, Chen Z. Progress and Prospects of Non-Canonical NF-κB Signaling Pathway in the Regulation of Liver Diseases. Molecules 2022; 27:molecules27134275. [PMID: 35807520 PMCID: PMC9268066 DOI: 10.3390/molecules27134275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 02/04/2023] Open
Abstract
Non-canonical nuclear factor kappa B (NF-κB) signaling pathway regulates many physiological and pathological processes, including liver homeostasis and diseases. Recent studies demonstrate that non-canonical NF-κB signaling pathway plays an essential role in hyperglycemia, non-alcoholic fatty liver disease, alcoholic liver disease, liver regeneration, liver injury, autoimmune liver disease, viral hepatitis, and hepatocellular carcinoma. Small-molecule inhibitors targeting to non-canonical NF-κB signaling pathway have been developed and shown promising results in the treatment of liver injuries. Here, the recent advances and future prospects in understanding the roles of the non-canonical NF-κB signaling pathways in the regulation of liver diseases are discussed.
Collapse
Affiliation(s)
- Li Tao
- Emergency Department, 305 Hospital of People’s Liberation Army, Beijing 100017, China; (L.T.); (W.Z.)
| | - Xiaomeng Ren
- College of Pharmaceutical and Biology Engineering, Shenyang University of Chemical Technology, Shenyang 110142, China
- Correspondence: (X.R.); (Z.C.); Tel.: +86-45186402029 (Z.C.)
| | - Wenhui Zhai
- Emergency Department, 305 Hospital of People’s Liberation Army, Beijing 100017, China; (L.T.); (W.Z.)
| | - Zheng Chen
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
- Correspondence: (X.R.); (Z.C.); Tel.: +86-45186402029 (Z.C.)
| |
Collapse
|
13
|
Amino acid sensor GCN2 promotes SARS-CoV-2 receptor ACE2 expression in response to amino acid deprivation. Commun Biol 2022; 5:651. [PMID: 35778545 PMCID: PMC9249868 DOI: 10.1038/s42003-022-03609-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 06/21/2022] [Indexed: 12/14/2022] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2) has been identified as a primary receptor for severe acute respiratory syndrome coronaviruses 2 (SARS-CoV-2). Here, we investigated the expression regulation of ACE2 in enterocytes under amino acid deprivation conditions. In this study, we found that ACE2 expression was upregulated upon all or single essential amino acid deprivation in human colonic epithelial CCD841 cells. Furthermore, we found that knockdown of general control nonderepressible 2 (GCN2) reduced intestinal ACE2 mRNA and protein levels in vitro and in vivo. Consistently, we revealed two GCN2 inhibitors, GCN2iB and GCN2-IN-1, downregulated ACE2 protein expression in CCD841 cells. Moreover, we found that increased ACE2 expression in response to leucine deprivation was GCN2 dependent. Through RNA-sequencing analysis, we identified two transcription factors, MAFB and MAFF, positively regulated ACE2 expression under leucine deprivation in CCD841 cells. These findings demonstrate that amino acid deficiency increases ACE2 expression and thereby likely aggravates intestinal SARS-CoV-2 infection. Amino acid deprivation increases ACE2 expression in the gut, potentially aggravating SARS-CoV-2 infection.
Collapse
|
14
|
Lan T, Qian S, Tang C, Gao J. Role of Immune Cells in Biliary Repair. Front Immunol 2022; 13:866040. [PMID: 35432349 PMCID: PMC9005827 DOI: 10.3389/fimmu.2022.866040] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/08/2022] [Indexed: 02/06/2023] Open
Abstract
The biliary system is comprised of cholangiocytes and plays an important role in maintaining liver function. Under normal conditions, cholangiocytes remain in the stationary phase and maintain a very low turnover rate. However, the robust biliary repair is initiated in disease conditions, and different repair mechanisms can be activated depending on the pathological changes. During biliary disease, immune cells including monocytes, lymphocytes, neutrophils, and mast cells are recruited to the liver. The cellular interactions between cholangiocytes and these recruited immune cells as well as hepatic resident immune cells, including Kupffer cells, determine disease outcomes. However, the role of immune cells in the initiation, regulation, and suspension of biliary repair remains elusive. The cellular processes of cholangiocyte proliferation, progenitor cell differentiation, and hepatocyte-cholangiocyte transdifferentiation during biliary diseases are reviewed to manifest the underlying mechanism of biliary repair. Furthermore, the potential role of immune cells in crucial biliary repair mechanisms is highlighted. The mechanisms of biliary repair in immune-mediated cholangiopathies, inherited cholangiopathies, obstructive cholangiopathies, and cholangiocarcinoma are also summarized. Additionally, novel techniques that could clarify the underlying mechanisms of biliary repair are displayed. Collectively, this review aims to deepen the understanding of the mechanisms of biliary repair and contributes potential novel therapeutic methods for treating biliary diseases.
Collapse
Affiliation(s)
- Tian Lan
- Lab of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Shuaijie Qian
- Lab of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Chengwei Tang
- Lab of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Jinhang Gao
- Lab of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
15
|
Li Z, Nie M, Yu L, Tao D, Wang Q, He Y, Liu Y, Zhang Y, Han H, Wang H. Blockade of the Notch Signaling Pathway Promotes M2 Macrophage Polarization to Suppress Cardiac Fibrosis Remodeling in Mice With Myocardial Infarction. Front Cardiovasc Med 2022; 8:639476. [PMID: 35111821 PMCID: PMC8801444 DOI: 10.3389/fcvm.2021.639476] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 11/24/2021] [Indexed: 11/30/2022] Open
Abstract
Myocardial infarction (MI) is regarded as a serious ischemic heart disease on a global level. The current study set out to explore the mechanism of the Notch signaling pathway in the regulation of fibrosis remodeling after the occurrence of MI. First, experimental mice were infected with recombination signal binding protein J (RBP-J) shRNA and empty adenovirus vector, followed by the establishment of MI mouse models and detection of cardiac function. After 4 weeks of MI, mice in the sh-RBP-J group were found to exhibit significantly improved cardiac function relative to the sh-NC group. Moreover, knockdown of RBP-J brought about decreased infarct area, promoted cardiac macrophages M2 polarization, reduced cardiac fibrosis, and further decreased transcription and protein expressions of inflammatory factors and fibrosis-related factors. Furthermore, downregulation of cylindromatosis (CYLD) using si-CYLD reversed the results that knockdown of RBP-J inhibited fibrogenesis and the release of inflammatory factors. Altogether, our findings indicated that the blockade of Notch signaling promotes M2 polarization of cardiac macrophages and improves cardiac function by inhibiting the imbalance of fibrotic remodeling after MI.
Collapse
Affiliation(s)
- Zhi Li
- Department of Cardiothoracic Surgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Miao Nie
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Liming Yu
- Department of Cardiothoracic Surgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Dengshun Tao
- Department of Cardiothoracic Surgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Qiang Wang
- Department of Cardiothoracic Surgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Yuanchen He
- Department of Cardiothoracic Surgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Yu Liu
- Department of Cardiothoracic Surgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Yuji Zhang
- Department of Cardiothoracic Surgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Hongguang Han
- Department of Cardiothoracic Surgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Huishan Wang
- Department of Cardiothoracic Surgery, General Hospital of Northern Theater Command, Shenyang, China
- *Correspondence: Huishan Wang
| |
Collapse
|
16
|
Ridder DA, Urbansky LL, Witzel HR, Schindeldecker M, Weinmann A, Berndt K, Gerber TS, Köhler BC, Nichetti F, Ludt A, Gehrke N, Schattenberg JM, Heinrich S, Roth W, Straub BK. Transforming Growth Factor-β Activated Kinase 1 (Tak1) Is Activated in Hepatocellular Carcinoma, Mediates Tumor Progression, and Predicts Unfavorable Outcome. Cancers (Basel) 2022; 14:cancers14020430. [PMID: 35053591 PMCID: PMC8774263 DOI: 10.3390/cancers14020430] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 01/12/2022] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Chronic inflammation is known to drive cancer initiation and progression in the liver and other organs. In different genetic mouse models, the role of the pro-inflammatory kinase Tak1 in liver cancer development has been controversial so far. To clarify the role of Tak1 in human hepatocellular carcinoma (HCC), we investigated the expression of Tak1 in a large and clinicopathologically well-characterized patient cohort with HCC. In human livers and HCCs, Tak1 is predominantly present in its isoform Tak1A localizing to the cell nucleus. Tak1 is upregulated in HCCs of the diethylnitrosamine mouse model as well as in human HCCs, independent of etiology, and is further induced in distant metastases. Overexpression of the isoform Tak1A in the HCC cell line Huh7 resulted in increased tumor cell migration, whereas overexpression of full-length Tak1 had no significant effect. In human HCCs, high nuclear Tak1 expression is associated with vascular invasion and short overall survival. Abstract Although knowledge on inflammatory signaling pathways driving cancer initiation and progression has been increasing, molecular mechanisms in hepatocarcinogenesis are still far from being completely understood. Hepatocyte-specific deletion of the MAPKKK Tak1 in mice recapitulates important steps of hepatocellular carcinoma (HCC) development, including the occurrence of cell death, steatohepatitis, dysplastic nodules, and HCCs. However, overactivation of Tak1 in mice upon deletion of its deubiquitinase Cyld also results in steatohepatitis and HCC development. To investigate Tak1 and Cyld in human HCCs, we created a tissue microarray to analyze their expression by immunohistochemistry in a large and well-characterized cohort of 871 HCCs of 561 patients. In the human liver and HCC, Tak1 is predominantly present as its isoform Tak1A and predominantly localizes to cell nuclei. Tak1 is upregulated in diethylnitrosamine-induced mouse HCCs as well as in human HCCs independent of etiology and is further induced in distant metastases. A high nuclear Tak1 expression is associated with short survival and vascular invasion. When we overexpressed Tak1A in Huh7 cells, we observed increased tumor cell migration, whereas overexpression of full-length Tak1 had no significant effect. A combined score of low Cyld and high Tak1 expression was an independent prognostic marker in a multivariate Cox regression model.
Collapse
Affiliation(s)
- Dirk Andreas Ridder
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (L.L.U.); (H.R.W.); (M.S.); (K.B.); (T.S.G.); (W.R.)
- Correspondence: (D.A.R.); (B.K.S.)
| | - Lana Louisa Urbansky
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (L.L.U.); (H.R.W.); (M.S.); (K.B.); (T.S.G.); (W.R.)
| | - Hagen Roland Witzel
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (L.L.U.); (H.R.W.); (M.S.); (K.B.); (T.S.G.); (W.R.)
| | - Mario Schindeldecker
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (L.L.U.); (H.R.W.); (M.S.); (K.B.); (T.S.G.); (W.R.)
- Tissue Biobank, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Arndt Weinmann
- Department of Internal Medicine, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (A.W.); (N.G.); (J.M.S.)
| | - Kristina Berndt
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (L.L.U.); (H.R.W.); (M.S.); (K.B.); (T.S.G.); (W.R.)
| | - Tiemo Sven Gerber
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (L.L.U.); (H.R.W.); (M.S.); (K.B.); (T.S.G.); (W.R.)
| | - Bruno Christian Köhler
- Department of Medical Oncology, National Center for Tumor Diseases, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Federico Nichetti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy;
- Computational Oncology, Molecular Diagnostics Program, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Annekathrin Ludt
- Institute of Medical Biostatistics, Epidemiology, and Informatics (IMBEI), University Medical Center Mainz, 55131 Mainz, Germany;
| | - Nadine Gehrke
- Department of Internal Medicine, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (A.W.); (N.G.); (J.M.S.)
| | - Jörn Markus Schattenberg
- Department of Internal Medicine, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (A.W.); (N.G.); (J.M.S.)
| | - Stefan Heinrich
- Department of General, Visceral and Transplant Surgery, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany;
| | - Wilfried Roth
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (L.L.U.); (H.R.W.); (M.S.); (K.B.); (T.S.G.); (W.R.)
| | - Beate Katharina Straub
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (L.L.U.); (H.R.W.); (M.S.); (K.B.); (T.S.G.); (W.R.)
- Correspondence: (D.A.R.); (B.K.S.)
| |
Collapse
|
17
|
Zhang J, Liu Q, He J, Li Y. Novel Therapeutic Targets in Liver Fibrosis. Front Mol Biosci 2021; 8:766855. [PMID: 34805276 PMCID: PMC8602792 DOI: 10.3389/fmolb.2021.766855] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/18/2021] [Indexed: 02/05/2023] Open
Abstract
Liver fibrosis is end-stage liver disease that can be rescued. If irritation continues due to viral infection, schistosomiasis and alcoholism, liver fibrosis can progress to liver cirrhosis and even cancer. The US Food and Drug Administration has not approved any drugs that act directly against liver fibrosis. The only treatments currently available are drugs that eliminate pathogenic factors, which show poor efficacy; and liver transplantation, which is expensive. This highlights the importance of clarifying the mechanism of liver fibrosis and searching for new treatments against it. This review summarizes how parenchymal, nonparenchymal cells, inflammatory cells and various processes (liver fibrosis, hepatic stellate cell activation, cell death and proliferation, deposition of extracellular matrix, cell metabolism, inflammation and epigenetics) contribute to liver fibrosis. We highlight discoveries of novel therapeutic targets, which may provide new insights into potential treatments for liver fibrosis.
Collapse
Affiliation(s)
- Jinhang Zhang
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Sichuan, China
| | - Qinhui Liu
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Sichuan, China
| | - Jinhan He
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Sichuan, China.,Department of Pharmacy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, China
| | - Yanping Li
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Sichuan, China
| |
Collapse
|
18
|
Brochado Ó, Martínez I, Berenguer J, Medrano L, González-García J, Jiménez-Sousa MÁ, Carrero A, Hontañón V, Navarro J, Guardiola JM, Fernández-Rodríguez A, Resino S. HCV eradication with IFN-based therapy does not completely restore gene expression in PBMCs from HIV/HCV-coinfected patients. J Biomed Sci 2021; 28:23. [PMID: 33785040 PMCID: PMC8010945 DOI: 10.1186/s12929-021-00718-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/24/2021] [Indexed: 12/11/2022] Open
Abstract
Objective To evaluate the impact of hepatitis C virus (HCV) elimination via interferon (IFN)-based therapy on gene expression profiles related to the immune system in HIV/HCV-coinfected patients. Methods We conducted a prospective study in 28 HIV/HCV-coinfected patients receiving IFN-based therapy at baseline (HIV/HCV-b) and week 24 after sustained virological response (HIV/HCV-f). Twenty-seven HIV-monoinfected patients (HIV-mono) were included as a control. RNA-seq analysis was performed on peripheral blood mononuclear cells (PBMCs). Genes with a fold-change (FC) ≥ 1.5 (in either direction) and false discovery rate (FDR) ≤ 0.05 were identified as significantly differentially expressed (SDE). Results HIV/HCV-b showed six SDE genes compared to HIV-mono group, but no significantly enriched pathways were observed. For HIV/HCV-f vs. HIV/HCV-b, we found 58 SDE genes, 34 upregulated and 24 downregulated in the HIV/HCV-f group. Of these, the most overexpressed were CXCL2, PDCD6IP, ATP5B, IGSF9, RAB26, and CSRNP1, and the most downregulated were IFI44 and IFI44L. These 58 SDE genes revealed two significantly enriched pathways (FDR < 0.05), one linked to Epstein-Barr virus infection and another related to p53 signaling. For HIV/HCV-f vs. HIV-mono group, we found 44 SDE genes that revealed 31 enriched pathways (FDR < 0.05) related to inflammation, cancer/cell cycle alteration, viral and bacterial infection, and comorbidities associated with HIV/HCV-coinfection. Five genes were overrepresented in most pathways (JUN, NFKBIA, PIK3R2, CDC42, and STAT3). Conclusion HIV/HCV-coinfected patients who eradicated hepatitis C with IFN-based therapy showed profound gene expression changes after achieving sustained virological response. The altered pathways were related to inflammation and liver-related complications, such as non-alcoholic fatty liver disease and hepatocellular carcinoma, underscoring the need for active surveillance for these patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12929-021-00718-6.
Collapse
Affiliation(s)
- Óscar Brochado
- Unidad de Infección Viral E Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III (Campus Majadahonda), Carretera Majadahonda-Pozuelo, Km 2.2, 28220, MajadahondaMadrid, Spain
| | - Isidoro Martínez
- Unidad de Infección Viral E Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III (Campus Majadahonda), Carretera Majadahonda-Pozuelo, Km 2.2, 28220, MajadahondaMadrid, Spain.
| | - Juan Berenguer
- Unidad de Enfermedades Infecciosas/VIH, Hospital General Universitario "Gregorio Marañón", Madrid, Spain.,Instituto de Investigación Sanitaria del Gregorio Marañón, Madrid, Spain
| | - Luz Medrano
- Unidad de Infección Viral E Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III (Campus Majadahonda), Carretera Majadahonda-Pozuelo, Km 2.2, 28220, MajadahondaMadrid, Spain
| | - Juan González-García
- Unidad de VIH, Servicio de Medicina Interna, Hospital Universitario "La Paz", Madrid, Spain.,Instituto de Investigacion Sanitaria La Paz (IdiPAZ), Madrid, Spain
| | - María Ángeles Jiménez-Sousa
- Unidad de Infección Viral E Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III (Campus Majadahonda), Carretera Majadahonda-Pozuelo, Km 2.2, 28220, MajadahondaMadrid, Spain
| | - Ana Carrero
- Unidad de Enfermedades Infecciosas/VIH, Hospital General Universitario "Gregorio Marañón", Madrid, Spain.,Instituto de Investigación Sanitaria del Gregorio Marañón, Madrid, Spain
| | - Víctor Hontañón
- Unidad de VIH, Servicio de Medicina Interna, Hospital Universitario "La Paz", Madrid, Spain.,Instituto de Investigacion Sanitaria La Paz (IdiPAZ), Madrid, Spain
| | - Jordi Navarro
- Servicio de Enfermedades Infecciosas, Hospital Universitari Vall D'Hebron, Barcelona, Spain.,Institut de Recerca Vall D'Hebron, Barcelona, Spain
| | | | - Amanda Fernández-Rodríguez
- Unidad de Infección Viral E Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III (Campus Majadahonda), Carretera Majadahonda-Pozuelo, Km 2.2, 28220, MajadahondaMadrid, Spain
| | - Salvador Resino
- Unidad de Infección Viral E Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III (Campus Majadahonda), Carretera Majadahonda-Pozuelo, Km 2.2, 28220, MajadahondaMadrid, Spain.
| | | |
Collapse
|
19
|
Chen Q, Lu X, Zhang X. Noncanonical NF-κB Signaling Pathway in Liver Diseases. J Clin Transl Hepatol 2021; 9:81-89. [PMID: 33604258 PMCID: PMC7868705 DOI: 10.14218/jcth.2020.00063] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 11/04/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022] Open
Abstract
The noncanonical NF-κB signaling pathway is an important branch of NF-κB signaling. It is involved in regulating multiple important biological processes, including inflammation and host immune response. A central adaptor protein of the noncanonical NF-κB pathway is NF-κB-inducing kinase (NIK), which activates the downstream kinase IKKα to process p100 to p52, thereby forming the RelB/p52 heterodimer to initiate the expression of target genes. Currently, many specific inhibitors and monoclonal antibodies targeting or triggering this pathway are being developed and tested for various diseases, including cancers, autoimmune diseases, and virus infection. Given that aberrant activation of the noncanonical NF-κB pathway is frequently observed in various liver diseases, targeting this pathway may be a promising therapeutic strategy to alleviate liver inflammation. Moreover, activation of this pathway may contribute to the antiviral immune response and promote the clearance of persistent hepatotropic virus infection. Here, we review the role of the noncanonical NF-κB pathway in the occurrence and development of different liver diseases, and discuss the potency and application of modulating the noncanonical NF-κB pathway for treatment of these liver diseases.
Collapse
Affiliation(s)
- Qianhui Chen
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou,Guangdong, China
| | - Xinyu Lu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou,Guangdong, China
| | - Xiaoyong Zhang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou,Guangdong, China
- Hepatology Unit, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
- Correspondence to: Xiaoyong Zhang, Hepatology Unit and Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, No. 1838, North Guangzhou Avenue, Guangzhou, Guangdong 510515, China. Tel: +86-20-62787830, E-mail:
| |
Collapse
|
20
|
Zhou D, Huang W, Wei J, Zhang J, Liu Z, Ji R, Ge S, Xiao M, Fan Y, Lu C. RelB promotes liver fibrosis via inducing the release of injury-associated inflammatory cytokines. J Cell Mol Med 2020; 24:6008-6014. [PMID: 32306539 PMCID: PMC7294124 DOI: 10.1111/jcmm.15108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/17/2020] [Accepted: 02/03/2020] [Indexed: 01/10/2023] Open
Abstract
Liver fibrosis is a serious chronic disease that developed by a coordinated interplay of many cell types, but the underlying signal transduction in individual cell type remains to be characterized. Nuclear factor‐κB (NF‐κB) is a widely accepted central player in the development of hepatic fibrosis. However, the precise role of each member of NF‐κB in different cell type is unclear. Here, we generated a mouse model (RelbΔhep) with hepatocyte‐specific deletion of RelB, a member of NF‐κB family. RelbΔhep mice born normally and appear normal without obvious abnormality. However, in the CCl4‐induced liver fibrosis, RelbΔhep mice developed less severe disease compared with wide‐type (WT) mice. The denaturation and necrosis of hepatocytes as well as the formation of false lobules in RelbΔhep mice were significantly reduced compared with WT mice. The production of α‐SMA and the level of collagen I and Collagen III were greatly reduced in RelbΔhep mice comparing with WT mice. Furthermore, in patients with liver fibrosis, RelB is up‐regulated along with the stage of diseases. Consistently, CCl4 treatment could up‐regulate the expression of RelB as well as inflammatory cytokines such as IL‐6 and TGF‐β1 in hepatoma cell as well as in WT mice. Knockdown the expression of RelB in hepatoma cells greatly reduced the expression of CCl4‐induced inflammatory cytokines. In summary, we provide the genetic evidence to demonstrate the critical and hepatocellular role of RelB in liver fibrosis. RelB is an important transcription factor to drive the expression of inflammatory cytokines in the initiation phase of injury.
Collapse
Affiliation(s)
- Danhua Zhou
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Jiangsu, China
| | - Wei Huang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Jiangsu, China
| | - Jinhuan Wei
- Laboratory of Medical Science, School of Medicine, Nantong University, Jiangsu, China
| | - Jingxin Zhang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Jiangsu, China
| | - Zhaoxiu Liu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Jiangsu, China
| | - Ran Ji
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Jiangsu, China
| | - Sijia Ge
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Jiangsu, China
| | - Mingbing Xiao
- Department of Gastroenterology and Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Yihui Fan
- Laboratory of Medical Science, School of Medicine, Nantong University, Jiangsu, China.,Department of Pathogenic Biology, School of Medicine, Nantong University, Jiangsu, China
| | - Cuihua Lu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Jiangsu, China
| |
Collapse
|
21
|
Guicciardi ME, Trussoni CE, LaRusso NF, Gores GJ. The Spectrum of Reactive Cholangiocytes in Primary Sclerosing Cholangitis. Hepatology 2020; 71:741-748. [PMID: 31833071 PMCID: PMC7012677 DOI: 10.1002/hep.31067] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 11/26/2019] [Indexed: 12/12/2022]
Abstract
Cholangiocytes are the target of a group of chronic liver diseases termed the "cholangiopathies," in which cholangiocytes react to exogenous and endogenous insults, leading to disease initiation and progression. In primary sclerosing cholangitis (PSC), the focus of this review, the cholangiocyte response to genetic or environmental insults can lead to a heterogeneous response; that is, a subpopulation acquires a ductular reactive and proliferative phenotype, while another subpopulation undergoes senescence and growth arrest. Both ductular reactive cholangiocytes and senescent cholangiocytes can modify the periductal microenvironment through their ability to secrete various cytokines, chemokines, and growth factors, initiating and perpetuating inflammatory and profibrotic responses. This review discusses the similarities and differences, the interrelationships, and the potential pathogenic roles of these reactive proliferative and senescent cholangiocyte subpopulations in PSC.
Collapse
Affiliation(s)
| | | | | | - Gregory J. Gores
- Corresponding author: Gregory J. Gores, MD., Professor of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, Tel: (507) 284-0686; Fax: (507) 284 0762;
| |
Collapse
|
22
|
Ko S, Russell JO, Molina LM, Monga SP. Liver Progenitors and Adult Cell Plasticity in Hepatic Injury and Repair: Knowns and Unknowns. ANNUAL REVIEW OF PATHOLOGY 2020; 15:23-50. [PMID: 31399003 PMCID: PMC7212705 DOI: 10.1146/annurev-pathmechdis-012419-032824] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The liver is a complex organ performing numerous vital physiological functions. For that reason, it possesses immense regenerative potential. The capacity for repair is largely attributable to the ability of its differentiated epithelial cells, hepatocytes and biliary epithelial cells, to proliferate after injury. However, in cases of extreme acute injury or prolonged chronic insult, the liver may fail to regenerate or do so suboptimally. This often results in life-threatening end-stage liver disease for which liver transplantation is the only effective treatment. In many forms of liver injury, bipotent liver progenitor cells are theorized to be activated as an additional tier of liver repair. However, the existence, origin, fate, activation, and contribution to regeneration of liver progenitor cells is hotly debated, especially since hepatocytes and biliary epithelial cells themselves may serve as facultative stem cells for one another during severe liver injury. Here, we discuss the evidence both supporting and refuting the existence of liver progenitor cells in a variety of experimental models. We also debate the validity of developing therapies harnessing the capabilities of these cells as potential treatments for patients with severe and chronic liver diseases.
Collapse
Affiliation(s)
- Sungjin Ko
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA;
- Pittsburgh Liver Research Center, University of Pittsburgh Medical Center and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| | - Jacquelyn O Russell
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA;
- Pittsburgh Liver Research Center, University of Pittsburgh Medical Center and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| | - Laura M Molina
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA;
- Pittsburgh Liver Research Center, University of Pittsburgh Medical Center and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| | - Satdarshan P Monga
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA;
- Pittsburgh Liver Research Center, University of Pittsburgh Medical Center and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| |
Collapse
|