1
|
Baek EB, Eun HS, Song JY, Hong EJ, Park SH, Kumbukgahadeniya P, Park SM, Kim SH, Kim SO, Kim HN, Cho YE, Won YS, Kwon HJ. Vitamin D supplementation ameliorates ductular reaction, liver inflammation and fibrosis in mice by upregulating TXNIP in ductular cells. Nat Commun 2025; 16:4420. [PMID: 40360509 PMCID: PMC12075793 DOI: 10.1038/s41467-025-59724-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 05/02/2025] [Indexed: 05/15/2025] Open
Abstract
Ductular reaction is associated with liver disease progression, but there are no drugs targeting ductular reaction. Vitamin D deficiency is common in chronic liver diseases and related to disease progression, but the underlying mechanisms by which vitamin D regulates liver diseases progression remain unclear. Here, we show that vitamin D plasma levels are negatively correlated with the degree of ductular reaction in patients with chronic liver diseases. 1,25(OH)2D3, the active form of vitamin D, reduces 3,5-diethoxycarbonyl-1,4-dihydrocollidin (DDC)-induced ductular reaction, liver inflammation, and fibrosis in female mice and upregulates the vitamin D target gene, TXNIP (encoding thioredoxin-interacting protein), in ductular cells. Cholangiocyte-specific Txnip-knockout female mice are more susceptible to DDC-induced ductular reaction, inflammation, and fibrosis. Deletion of Txnip in cholangiocytes promotes proliferation and suppressed death. Furthermore, Txnip deficiency increases TNF-α and TGF-β secretion by cholangiocytes to stimulate Kupffer cells and hepatic stellate cells, consequently leading to inflammation and collagen deposition. Biliary Txnip deficiency abolishes the protective effects of vitamin D, and TXNIP overexpression attenuates DDC-induced ductular reaction and inflammation and fibrosis. Collectively, our findings identify new mechanism how vitamin D ameliorates liver diseases and suggest that the vitamin D/TXNIP axis is a therapeutic target for addressing ductular reaction and liver diseases.
Collapse
Affiliation(s)
- Eun Bok Baek
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Physiology, College of Medicine, Jeju National University, Jeju, Republic of Korea
| | - Hyuk Soo Eun
- Department of Internal Medicine, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Internal Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Jun-Yeop Song
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Eun-Ju Hong
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Se-Hee Park
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Chungbuk, Republic of Korea
| | | | - Sang-Min Park
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Seok-Hwan Kim
- Department of Surgery, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Soon Ok Kim
- Department of Medical Sciences, Chungnam National University, Daejeon, Republic of Korea
| | - Ha Neul Kim
- Department of Medical Sciences, Chungnam National University, Daejeon, Republic of Korea
| | - Young-Eun Cho
- Department of Food and Nutrition, Andong National University, Andong, Republic of Korea
| | - Young-Suk Won
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Chungbuk, Republic of Korea.
| | - Hyo-Jung Kwon
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea.
| |
Collapse
|
2
|
Li Q, Meng Q, Niu N, Li Y, Lu W, Wang T, Li Y, Ju W, Ma Y, Wu J. Yinchenzhufu decoction ameliorates cholestatic liver injury through decreasing the formation of neutrophil extracellular traps in mice. JOURNAL OF ETHNOPHARMACOLOGY 2025; 347:119720. [PMID: 40174730 DOI: 10.1016/j.jep.2025.119720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 03/24/2025] [Accepted: 03/29/2025] [Indexed: 04/04/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Neutrophil extracellular traps (NETs) are important driving factors to liver disease. Yinchenzhufu decoction (YCZFD) is a classical traditional herbal medicine used to treat cholestatic liver injury (CLI); however, its effect on NETs in CLI remains unknown. AIM OF THE STUDY To elucidate the potential molecular mechanism of YCZFD against CLI through multiple omics techniques. MATERIALS AND METHODS The protective effects of YCZFD against cholestatic liver injury and fibrosis were investigated in a 3, 5-Diethoxycarbonyl-1, 4-Dihydro-2, 4, 6-Collidine (DDC) induced cholestatic mouse model. Its effects on blood biochemical indicators, liver tissue morphology, neutrophil infiltration, and fibrosis in cholestatic mice were determined. Then RNA sequencing, quantitative proteomics, and molecular biology were used to analyze the mechanism of YCZFD against CLI. RESULTS Treatment different doses of YCZFD significantly ameliorated neutrophil infiltration, alleviating CLI in cholestatic mice. RNA sequencing showed that neutrophil recruitment genes, such as CXCL5 and CXCR2, were upregulated in the liver tissues of cholestatic mice, but YCZFD treatment decreased this expression. Additionally, quantitative proteomics revealed that liver myeloperoxidase and S100A9 expression, key marker proteins of NETs, was significantly increased in cholestatic mice and decreased by YCZFD. Similarly, molecular biology confirmed that YCZFD reduced NETs proteins expression and ameliorated NETs formation in the liver of cholestatic mice. As NETs promotes inflammation, further results showed that YCZFD treatment inhibited the NF-κB signaling pathway and collagen formation, alleviating liver inflammation and fibrosis of cholestatic mice. CONCLUSIONS YCZFD decreases neutrophil recruitment and formation of NETs, in turn alleviating cholestatic liver injury and fibrosis.
Collapse
Affiliation(s)
- Qi Li
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qian Meng
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ningning Niu
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiqian Li
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenye Lu
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tianming Wang
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuanyuan Li
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Wanjun Ju
- Department of Endocrinology, Shanghai Pudong New Area Hospital of Traditional Chinese Medicine, Shanghai, China.
| | - Yueming Ma
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Jiasheng Wu
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
3
|
Arias E, Haynes ME, Nadkarni NA, Lipfert ZK, Muller WA, Batra A, Sullivan DP. EdU tracking of leukocyte recruitment in mouse models of ischemic stroke and sterile lung inflammation. J Cell Sci 2025; 138:jcs263835. [PMID: 40260638 DOI: 10.1242/jcs.263835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/17/2025] [Indexed: 04/23/2025] Open
Abstract
The discovery of copper(I)-catalyzed azide-alkyne cycloaddition (click chemistry) has significantly advanced the detection of proliferating cells by utilizing 5-ethynyl-2'-deoxyuridine (EdU). EdU, a thymidine analogue, is incorporated into DNA during replication and detected by the direct reaction with an azide-conjugated fluorophore. Traditionally, dividing cells are labeled using 5-bromodeoxyuridine (BrdU), another nucleotide analogue. However, BrdU detection is a harsh method that requires substantial sample processing, unlike EdU detection. EdU is classically used to identify proliferating cells; however, we report a streamlined methodology that uses EdU to label and track leukocyte recruitment that is compatible with flow cytometry and microscopy and preserves transgenic fluorophores. EdU labeling was performed in two different models of sterile inflammation: ischemic stroke and hydrochloric acid aspiration. EdU injection was timed to differentially label circulating monocytes, neutrophils and T cells. Tissue analysis showed EdU-positive monocytes and T cells were enriched in both inflammatory models. This suggests that recently divided monocytes and T cells are preferentially recruited to these vascular beds during inflammation and highlights the utility of this labeling approach to track leukocyte subtypes longitudinally during inflammation.
Collapse
Affiliation(s)
- Erika Arias
- Department of Pathology, Northwestern University, Chicago, IL 60643, USA
| | - Maureen E Haynes
- Department of Pathology, Northwestern University, Chicago, IL 60643, USA
| | - Neil A Nadkarni
- Department of Neurology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Zoie K Lipfert
- Department of Neurology, Northwestern University, Chicago, IL 60643, USA
| | - William A Muller
- Department of Pathology, Northwestern University, Chicago, IL 60643, USA
| | - Ayush Batra
- Department of Pathology, Northwestern University, Chicago, IL 60643, USA
- Department of Neurology, Northwestern University, Chicago, IL 60643, USA
| | - David P Sullivan
- Department of Pathology, Northwestern University, Chicago, IL 60643, USA
| |
Collapse
|
4
|
Gupta V, Sehrawat TS, Pinzani M, Strazzabosco M. Portal Fibrosis and the Ductular Reaction: Pathophysiological Role in the Progression of Liver Disease and Translational Opportunities. Gastroenterology 2025; 168:675-690. [PMID: 39251168 PMCID: PMC11885590 DOI: 10.1053/j.gastro.2024.07.044] [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: 03/18/2024] [Revised: 06/27/2024] [Accepted: 07/20/2024] [Indexed: 09/11/2024]
Abstract
A consistent feature of chronic liver diseases and the hallmark of pathologic repair is the so-called "ductular reaction." This is a histologic abnormality characterized by an expansion of dysmorphic cholangiocytes inside and around portal spaces infiltrated by inflammatory, mesenchymal, and vascular cells. The ductular reaction is a highly regulated response based on the reactivation of morphogenetic signaling mechanisms and a complex crosstalk among a multitude of cell types. The nature and mechanism of these exchanges determine the difference between healthy regenerative liver repair and pathologic repair. An orchestrated signaling among cell types directs mesenchymal cells to deposit a specific extracellular matrix with distinct physical and biochemical properties defined as portal fibrosis. Progression of fibrosis leads to vast architectural and vascular changes known as "liver cirrhosis." The signals regulating the ecology of this microenvironment are just beginning to be addressed. Contrary to the tumor microenvironment, immune modulation inside this "benign" microenvironment is scarcely known. One of the reasons for this is that both the ductular reaction and portal fibrosis have been primarily considered a manifestation of cholestatic liver disease, whereas this phenomenon is also present, albeit with distinctive features, in all chronic human liver diseases. Novel human-derived cellular models and progress in "omics" technologies are increasing our knowledge at a fast pace. Most importantly, this knowledge is on the edge of generating new diagnostic and therapeutic advances. Here, we will critically review the latest advances, in terms of mechanisms, pathophysiology, and treatment prospects. In addition, we will delineate future avenues of research, including innovative translational opportunities.
Collapse
Affiliation(s)
- Vikas Gupta
- Liver Center and Section of Digestive Diseases, Department of Internal Medicine, Yale University, New Haven, Connecticut
| | - Tejasav S Sehrawat
- Liver Center and Section of Digestive Diseases, Department of Internal Medicine, Yale University, New Haven, Connecticut
| | - Massimo Pinzani
- UCL Institute for Liver & Digestive Health, Royal Free Hospital, London, United Kingdom; University of Pittsburgh Medical Center-Mediterranean Institute for Transplantation and Highly Specialized Therapies, Palermo, Italy
| | - Mario Strazzabosco
- Liver Center and Section of Digestive Diseases, Department of Internal Medicine, Yale University, New Haven, Connecticut.
| |
Collapse
|
5
|
Gan C, Yuan Y, Shen H, Gao J, Kong X, Che Z, Guo Y, Wang H, Dong E, Xiao J. Liver diseases: epidemiology, causes, trends and predictions. Signal Transduct Target Ther 2025; 10:33. [PMID: 39904973 PMCID: PMC11794951 DOI: 10.1038/s41392-024-02072-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 10/06/2024] [Accepted: 11/12/2024] [Indexed: 02/06/2025] Open
Abstract
As a highly complex organ with digestive, endocrine, and immune-regulatory functions, the liver is pivotal in maintaining physiological homeostasis through its roles in metabolism, detoxification, and immune response. Various factors including viruses, alcohol, metabolites, toxins, and other pathogenic agents can compromise liver function, leading to acute or chronic injury that may progress to end-stage liver diseases. While sharing common features, liver diseases exhibit distinct pathophysiological, clinical, and therapeutic profiles. Currently, liver diseases contribute to approximately 2 million deaths globally each year, imposing significant economic and social burdens worldwide. However, there is no cure for many kinds of liver diseases, partly due to a lack of thorough understanding of the development of these liver diseases. Therefore, this review provides a comprehensive examination of the epidemiology and characteristics of liver diseases, covering a spectrum from acute and chronic conditions to end-stage manifestations. We also highlight the multifaceted mechanisms underlying the initiation and progression of liver diseases, spanning molecular and cellular levels to organ networks. Additionally, this review offers updates on innovative diagnostic techniques, current treatments, and potential therapeutic targets presently under clinical evaluation. Recent advances in understanding the pathogenesis of liver diseases hold critical implications and translational value for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Can Gan
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuan Yuan
- Aier Institute of Ophthalmology, Central South University, Changsha, China
| | - Haiyuan Shen
- Department of Oncology, the First Affiliated Hospital; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Jinhang Gao
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiangxin Kong
- Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Zhaodi Che
- Clinical Medicine Research Institute and Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yangkun Guo
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China.
| | - Erdan Dong
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China.
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
| | - Jia Xiao
- Clinical Medicine Research Institute and Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou, China.
- Department of Gastroenterology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, China.
| |
Collapse
|
6
|
Greenman R, Weston CJ. CCL24 and Fibrosis: A Narrative Review of Existing Evidence and Mechanisms. Cells 2025; 14:105. [PMID: 39851534 PMCID: PMC11763828 DOI: 10.3390/cells14020105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 01/06/2025] [Accepted: 01/10/2025] [Indexed: 01/26/2025] Open
Abstract
Tissue fibrosis results from a dysregulated and chronic wound healing response accompanied by chronic inflammation and angiogenesis. Regardless of the affected organ, fibrosis shares the following common hallmarks: the recruitment of immune cells, fibroblast activation/proliferation, and excessive extracellular matrix deposition. Chemokines play a pivotal role in initiating and advancing these fibrotic processes. CCL24 (eotaxin-2) is a chemokine secreted by immune cells and epithelial cells, which promotes the trafficking of immune cells and the activation of profibrotic cells through CCR3 receptor binding. Higher levels of CCL24 and CCR3 were found in the tissue and sera of patients with fibro-inflammatory diseases, including primary sclerosing cholangitis (PSC), systemic sclerosis (SSc), and metabolic dysfunction-associated steatohepatitis (MASH). This review delves into the intricate role of CCL24 in fibrotic diseases, highlighting its impact on fibrotic, immune, and vascular pathways. We focus on the preclinical and clinical evidence supporting the therapeutic potential of blocking CCL24 in diseases that involve excessive inflammation and fibrosis.
Collapse
Affiliation(s)
| | - Chris J. Weston
- Department of Immunology and Immunotherapy, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
- National Institute for Health and Care Research (NIHR), Birmingham Biomedical Research Centre, Birmingham B15 2TT, UK
| |
Collapse
|
7
|
Tang S, Wu S, Zhang W, Ma L, Zuo L, Wang H. Immunology and treatments of fatty liver disease. Arch Toxicol 2025; 99:127-152. [PMID: 39692857 DOI: 10.1007/s00204-024-03920-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024]
Abstract
Alcoholic liver disease (ALD) and non-alcoholic fatty liver disease (NAFLD) are two major chronic liver diseases worldwide. The triggers for fatty liver can be derived from external sources such as adipose tissue, the gut, personal diet, and genetics, or internal sources, including immune cell responses, lipotoxicity, hepatocyte death, mitochondrial dysfunction, and extracellular vesicles. However, their pathogenesis varies to some extent. This review summarizes various immune mechanisms and therapeutic targets associated with these two types of fatty liver disease. It describes the gut-liver axis and adipose tissue-liver crosstalk, as well as the roles of different immune cells (both innate and adaptive immune cells) in fatty liver disease. Additionally, mitochondrial dysfunction, extracellular vesicles, microRNAs (miRNAs), and gastrointestinal hormones are also related to the pathogenesis of fatty liver. Understanding the pathogenesis of fatty liver and corresponding therapeutic strategies provides a new perspective for developing novel treatments for fatty liver disease.
Collapse
Affiliation(s)
- Sainan Tang
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China
- Laboratory of Molecular Biology, Department of Biochemistry, School of Basic Medical Science, Anhui Medical University, Hefei, 230022, Anhui, China
| | - Shanshan Wu
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China
- Department of Gastroenterology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, Anhui, China
| | - Wenzhe Zhang
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China
- The First College of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Lili Ma
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China
- The First College of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Li Zuo
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China.
- Laboratory of Molecular Biology, Department of Biochemistry, School of Basic Medical Science, Anhui Medical University, Hefei, 230022, Anhui, China.
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China.
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
8
|
Herro R, Grimes HL. The diverse roles of neutrophils from protection to pathogenesis. Nat Immunol 2024; 25:2209-2219. [PMID: 39567761 DOI: 10.1038/s41590-024-02006-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 10/07/2024] [Indexed: 11/22/2024]
Abstract
Neutrophil granulocytes are the most abundant leukocytes in the blood and constitute a critical arm of innate immunity. They are generated in the bone marrow, and under homeostatic conditions enter the bloodstream to patrol tissues and scout for potential pathogens that they quickly destroy through phagocytosis, intracellular degradation, release of granules and formation of extracellular traps. Thus, neutrophils are important effector cells involved in antibacterial defense. However, neutrophils can also be pathogenic. Emerging data suggest they have critical functions related to tissue repair and fibrosis. Moreover, similarly to other innate immune cells, neutrophil cell states are affected by their microenvironment. Notably, this includes tumors that co-opt neutrophils. Neutrophils can undergo transcriptional and epigenetic reprogramming, thus causing or modulating inflammation and injury. It is also possible that distinct neutrophil subsets are generated with designated functions in the bone marrow. Understanding neutrophil plasticity and alternative cell states will help resolve their contradictive roles. This Review summarizes the most recent key findings surrounding protective versus pathogenic functions of neutrophils; elaborating on phenotype-specific subsets of neutrophils and their involvement in homeostasis and disease.
Collapse
Affiliation(s)
- Rana Herro
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, USA.
| | - H Leighton Grimes
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
9
|
Alvarado-Tapias E, Maya-Miles D, Albillos A, Aller R, Ampuero J, Andrade RJ, Arechederra M, Aspichueta P, Banales JM, Blas-García A, Caparros E, Cardoso Delgado T, Carrillo-Vico A, Claria J, Cubero FJ, Díaz-Ruiz A, Fernández-Barrena MG, Fernández-Iglesias A, Fernández-Veledo S, Francés R, Gallego-Durán R, Gracia-Sancho J, Irimia M, Lens S, Martínez-Chantar ML, Mínguez B, Muñoz-Hernández R, Nogueiras R, Ramos-Molina B, Riveiro-Barciela M, Rodríguez-Perálvarez ML, Romero-Gómez M, Sabio G, Sancho-Bru P, Ventura-Cots M, Vidal S, Gahete MD. Proceedings of the 5th Meeting of Translational Hepatology, organized by the Spanish Association for the Study of the Liver (AEEH). GASTROENTEROLOGIA Y HEPATOLOGIA 2024; 47:502207. [PMID: 38723772 DOI: 10.1016/j.gastrohep.2024.502207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 05/02/2024] [Indexed: 11/30/2024]
Abstract
This is the summary report of the 5th Translational Hepatology Meeting, endorsed by the Spanish Association for the Study of the Liver (AEEH) and held in Seville, Spain, in October 2023. The meeting aimed to provide an update on the latest advances in the field of basic and translational hepatology, covering different molecular, cellular, and pathophysiological aspects of the most relevant clinical challenges in liver pathologies. This includes the identification of novel biomarkers and diagnostic tools, the understanding of the relevance of immune response and inflammation in liver diseases, the characterization of current medical approaches to reverse liver diseases, the incorporation of novel molecular insights through omics techniques, or the characterization of the impact of toxic and metabolic insults, as well as other organ crosstalk, in liver pathophysiology.
Collapse
Affiliation(s)
- Edilmar Alvarado-Tapias
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Gastroenterology, Hospital Santa Creu I Sant Pau, Institut de Recerca Sant Pau, Universidad Autónoma de Barcelona, Barcelona, Spain.
| | - Douglas Maya-Miles
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío (HUVR), CISC, Universidad de Sevilla, Sevilla, Spain.
| | - Agustin Albillos
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Servicio de Gastroenterología y Hepatología, Hospital Universitario Ramón y Cajal/Universidad de Alcalá/Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Rocio Aller
- BioCritic, Group for Biomedical Research in Critical Care Medicine, Spain; Department of Medicine, Dermatology and Toxicology, Universidad de Valladolid, Spain; Gastroenterology Unit, Hospital Clínico Universitario de Valladolid, 47003 Valladolid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga, Spain
| | - Javier Ampuero
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío (HUVR), CISC, Universidad de Sevilla, Sevilla, Spain
| | - Raul J Andrade
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga, Spain
| | - Maria Arechederra
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain; Instituto de Investigaciones Sanitarias de Navarra IdiSNA, Pamplona, Spain
| | - Patricia Aspichueta
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Spain; Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Jesus M Banales
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute - Donostia University Hospital - University of the Basque Country (UPV/EHU), Ikerbasque, Donostia-San Sebastian, Spain; Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Ana Blas-García
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Departamento de Fisiología, Universitat de València, Av. Blasco Ibáñez, 15, 46010 Valencia, Spain; FISABIO (Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana), Av. de Catalunya, 21, 46020 Valencia, Spain
| | - Esther Caparros
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Grupo de Inmunobiología Hepática e Intestinal, Departamento Medicina Clínica, Universidad Miguel Hernández, San Juan, Spain; Instituto de Investigación Sanitaria ISABIAL, Hospital General Universitario de Alicante, Alicante, Spain
| | - Teresa Cardoso Delgado
- Biobizkaia Health Research Institute, Barakaldo, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Antonio Carrillo-Vico
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío (HUVR), CISC, Universidad de Sevilla, Sevilla, Spain; Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| | - Joan Claria
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Biochemistry and Molecular Genetics Service, Hospital Clínic, IDIBAPS, Barcelona, Spain; University of Barcelona, Spain
| | - Francisco Javier Cubero
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain; Health Research Institute Gregorio Marañón (IiSGM), Madrid, Spain
| | - Alberto Díaz-Ruiz
- Laboratory of Cellular and Molecular Gerontology, Precision Nutrition and Aging, Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM+CSIC, Madrid, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Maite G Fernández-Barrena
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Investigaciones Sanitarias de Navarra IdiSNA, Pamplona, Spain; Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Spain
| | - Anabel Fernández-Iglesias
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Liver Vascular Biology Research Group, IDIBAPS, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - Sonia Fernández-Veledo
- Department of Endocrinology and Nutrition and Research Unit, University Hospital of Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), Tarragona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Ruben Francés
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Grupo de Inmunobiología Hepática e Intestinal, Departamento Medicina Clínica, Universidad Miguel Hernández, San Juan, Spain; Instituto de Investigación Sanitaria ISABIAL, Hospital General Universitario de Alicante, Alicante, Spain
| | - Rocío Gallego-Durán
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío (HUVR), CISC, Universidad de Sevilla, Sevilla, Spain
| | - Jordi Gracia-Sancho
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Liver Vascular Biology Research Group, IDIBAPS, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - Manuel Irimia
- Universitat Pompeu Fabra (UPF), Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, ICREA, Barcelona, Spain
| | - Sabela Lens
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain; Liver Unit, Hospital Clínic, IDIBAPS, Barcelona, Spain
| | - María Luz Martínez-Chantar
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia, Spain
| | - Beatriz Mínguez
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Liver Unit, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Rocío Muñoz-Hernández
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío (HUVR), CISC, Universidad de Sevilla, Sevilla, Spain; Departamento de fisiología, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain
| | - Rubén Nogueiras
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain; Department of Physiology, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain; Galician Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, Spain
| | - Bruno Ramos-Molina
- Obesity, Diabetes and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Mar Riveiro-Barciela
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Liver Unit, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Manuel L Rodríguez-Perálvarez
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Hepatology and Liver Transplantation, Reina Sofia University Hospital, Cordoba, Spain; Maimonides Biomedical Research Institute of Córdoba (IMIBIC), University of Córdoba, Cordoba, Spain
| | - Manuel Romero-Gómez
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío (HUVR), CISC, Universidad de Sevilla, Sevilla, Spain
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Stress Kinases in Diabetes, Cancer and Biochemistry, Spain; Centro Nacional de Investigaciones Oncologicas (CNIO), Organ Crosstalk in Metabolic Diseases, Madrid, Spain
| | - Pau Sancho-Bru
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Meritxell Ventura-Cots
- Liver Unit, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain; Center for Liver Diseases, Pittsburgh Liver Research Center, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Silvia Vidal
- Group of Inflammatory Diseases, Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Manuel D Gahete
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Spain; Molecular Hepatology Group, Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Spain; Reina Sofia University Hospital, Cordoba, Spain.
| |
Collapse
|
10
|
Stonebraker JR, Pace RG, Gallins PJ, Dang H, Aksit M, Faino AV, Gordon WW, MacParland S, Bamshad MJ, Gibson RL, Cutting GR, Durie PR, Wright FA, Zhou YH, Blackman SM, O’Neal WK, Ling SC, Knowles MR. Genetic variation in severe cystic fibrosis liver disease is associated with novel mechanisms for disease pathogenesis. Hepatology 2024; 80:1012-1025. [PMID: 38536042 PMCID: PMC11427593 DOI: 10.1097/hep.0000000000000863] [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: 12/15/2023] [Accepted: 03/11/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND AND AIMS It is not known why severe cystic fibrosis (CF) liver disease (CFLD) with portal hypertension occurs in only ~7% of people with CF. We aimed to identify genetic modifiers for severe CFLD to improve understanding of disease mechanisms. APPROACH AND RESULTS Whole-genome sequencing was available in 4082 people with CF with pancreatic insufficiency (n = 516 with severe CFLD; n = 3566 without CFLD). We tested ~15.9 million single nucleotide polymorphisms (SNPs) for association with severe CFLD versus no-CFLD, using pre-modulator clinical phenotypes including (1) genetic variant ( SERPINA1 ; Z allele) previously associated with severe CFLD; (2) candidate SNPs (n = 205) associated with non-CF liver diseases; (3) genome-wide association study of common/rare SNPs; (4) transcriptome-wide association; and (5) gene-level and pathway analyses. The Z allele was significantly associated with severe CFLD ( p = 1.1 × 10 -4 ). No significant candidate SNPs were identified. A genome-wide association study identified genome-wide significant SNPs in 2 loci and 2 suggestive loci. These 4 loci contained genes [significant, PKD1 ( p = 8.05 × 10 -10 ) and FNBP1 ( p = 4.74 × 10 -9 ); suggestive, DUSP6 ( p = 1.51 × 10 -7 ) and ANKUB1 ( p = 4.69 × 10 -7 )] relevant to severe CFLD pathophysiology. The transcriptome-wide association identified 3 genes [ CXCR1 ( p = 1.01 × 10 -6 ) , AAMP ( p = 1.07 × 10 -6 ), and TRBV24 ( p = 1.23 × 10 -5 )] involved in hepatic inflammation and innate immunity. Gene-ranked analyses identified pathways enriched in genes linked to multiple liver pathologies. CONCLUSION These results identify loci/genes associated with severe CFLD that point to disease mechanisms involving hepatic fibrosis, inflammation, innate immune function, vascular pathology, intracellular signaling, actin cytoskeleton and tight junction integrity and mechanisms of hepatic steatosis and insulin resistance. These discoveries will facilitate mechanistic studies and the development of therapeutics for severe CFLD.
Collapse
Affiliation(s)
- Jaclyn R. Stonebraker
- Marsico Lung Institute/UNC CF Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA
| | - Rhonda G. Pace
- Marsico Lung Institute/UNC CF Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA
| | - Paul J. Gallins
- Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina, 27695, USA
| | - Hong Dang
- Marsico Lung Institute/UNC CF Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA
| | - M.A. Aksit
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA
| | - Anna V. Faino
- Children’s Core for Biostatistics, Epidemiology and Analytics in Research, Seattle Children’s Research Institute, Seattle, Washington, 98101, USA
| | - William W. Gordon
- Department of Pediatrics, Division of Genetic Medicine, University of Washington, Seattle, Washington, 98195, USA
| | - Sonya MacParland
- Ajmera Transplant Centre, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Michael J. Bamshad
- Department of Pediatrics, Division of Genetic Medicine, University of Washington, Seattle, Washington, 98195, USA
- Brotman Baty Institute for Precision Medicine, Seattle, Washington, 98195, USA
- Department of Genome Sciences, University of Washington, Seattle, Washington, 98195, USA
| | - Ronald L. Gibson
- Center for Respiratory Biology & Therapeutics, Seattle Children’s Research Institute, Seattle, Washington, 98105, USA
| | - Garry R. Cutting
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA
| | | | - Fred A. Wright
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, 27695, USA
- Department of Statistics, North Carolina State University, Raleigh, North Carolina, 27695, USA
| | - Yi-Hui Zhou
- Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina, 27695, USA
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, 27695, USA
| | - Scott M. Blackman
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA
- Division of Pediatric Endocrinology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA
| | - Wanda K. O’Neal
- Marsico Lung Institute/UNC CF Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA
| | - Simon C. Ling
- Division of Gastroenterology, Hepatology, and Nutrition, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Michael R. Knowles
- Marsico Lung Institute/UNC CF Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA
| |
Collapse
|
11
|
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
|
12
|
Cornillet M, Geanon D, Bergquist A, Björkström NK. Immunobiology of primary sclerosing cholangitis. Hepatology 2024:01515467-990000000-01014. [PMID: 39226402 DOI: 10.1097/hep.0000000000001080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 08/21/2024] [Indexed: 09/05/2024]
Abstract
Primary sclerosing cholangitis (PSC) is a chronic inflammatory progressive cholestatic liver disease. Genetic risk factors, the presence of autoantibodies, the strong clinical link with inflammatory bowel disease, and associations with other autoimmune disorders all suggest a pivotal role for the immune system in PSC pathogenesis. In this review, we provide a comprehensive overview of recent immunobiology insights in PSC. A particular emphasis is given to immunological concepts such as tissue residency and knowledge gained from novel technologies, including single-cell RNA sequencing and spatial transcriptomics. This review of the immunobiological landscape of PSC covers major immune cell types known to be enriched in PSC-diseased livers as well as recently described cell types whose biliary localization and contribution to PSC immunopathogenesis remain incompletely described. Finally, we emphasize the importance of time and space in relation to PSC heterogeneity as a key consideration for future studies interrogating the role of the immune system in PSC.
Collapse
Affiliation(s)
- Martin Cornillet
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Daniel Geanon
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Annika Bergquist
- Unit of Gastroenterology, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Niklas K Björkström
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
13
|
Sun J, Yang F, Zheng Y, Huang C, Fan X, Yang L. Pathogenesis and interaction of neutrophils and extracellular vesicles in noncancer liver diseases. Int Immunopharmacol 2024; 137:112442. [PMID: 38889508 DOI: 10.1016/j.intimp.2024.112442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/20/2024]
Abstract
Liver disease ranks as the eleventh leading cause of mortality, leading to approximately 2 million deaths annually worldwide. Neutrophils are a type of immune cell that are abundant in peripheral blood and play a vital role in innate immunity by quickly reaching the site of liver injury. They exert their influence on liver diseases through autocrine, paracrine, and immunomodulatory mechanisms. Extracellular vesicles, phospholipid bilayer vesicles, transport a variety of substances, such as proteins, nucleic acids, lipids, and pathogenic factors, for intercellular communication. They regulate cell communication and perform their functions by delivering biological information. Current research has revealed the involvement of the interaction between neutrophils and extracellular vesicles in the pathogenesis of liver disease. Moreover, more research has focused on targeting neutrophils as a therapeutic strategy to attenuate disease progression. Therefore, this article summarizes the roles of neutrophils, extracellular vesicles, and their interactions in noncancerous liver diseases.
Collapse
Affiliation(s)
- Jie Sun
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital, Sichuan University, Chengdu, China; Medical College, Tibet University, Lhasa, China
| | - Fan Yang
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Yanyi Zheng
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Chen Huang
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoli Fan
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital, Sichuan University, Chengdu, China.
| | - Li Yang
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
14
|
Wang Y, Liu X, Li K, Wang X, Zhang X, Qian D, Meng X, Yu L, Yan X, He Z. Self-Sulfhydrated, Nitro-Fixed Albumin Nanoparticles as a Potent Therapeutic Agent for the Treatment of Acute Liver Injury. ACS NANO 2024. [PMID: 39041805 DOI: 10.1021/acsnano.4c07297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Exogenous polysulfhydryls (R-SH) supplementation and nitric oxide (NO) gas molecules delivery provide essential antioxidant buffering pool components and anti-inflammatory species in cellular defense against injury, respectively. Herein, the intermolecular disulfide bonds in bovine serum albumin (BSA) molecules were reductively cleaved under native and mild conditions to expose multiple sulfhydryl groups (BSA-SH), then sulfhydryl-nitrosylated (R-SNO), and nanoprecipitated to form injectable self-sulfhydrated, nitro-fixed albumin nanoparticles (BSA-SNO NPs), allowing albumin to act as a NO donor reservoir and multiple sulfhydryl group transporter while also preventing unfavorable oxidation and self-cross-linking of polysulfhydryl groups. In two mouse models of ischemia/reperfusion-induced and endotoxin-induced acute liver injury (ALI), a single low dosage of BSA-SNO NPs (S-nitrosothiols: 4 μmol·kg-1) effectively attenuated oxidative stress and systemic inflammation cascades in the upstream pathophysiology of disease progression, thus rescuing dying hepatocytes, regulating host defense, repairing microcirculation, and restoring liver function. By mechanistically upregulating the antioxidative signaling pathway (Nrf-2/HO-1/NOQ1) and inhibiting the inflammatory cytokine storm (NF-κB/p-IκBα/TNF-α/IL-β), BSA-SNO NPs blocked the initiation of the mitochondrial apoptotic signaling pathway (Cyto C/Bcl-2 family/caspase-3) and downregulated the cell pyroptosis pathway (NLRP3/ASC/IL-1β), resulting in an increased survival rate from 26.7 to 73.3%. This self-sulfhydrated, nitro-fixed functionalized BSA nanoformulation proposes a potential drug-free treatment strategy for ALI.
Collapse
Affiliation(s)
- Yanan Wang
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao/Sanya 266100/572024, China
- Sanya Oceanographic Institution, Sanya 572024, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao 266003, China
| | - Xiaohu Liu
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao/Sanya 266100/572024, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao 266003, China
| | - Keyang Li
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao/Sanya 266100/572024, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao 266003, China
| | - Xinyuan Wang
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao/Sanya 266100/572024, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao 266003, China
| | - Xue Zhang
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao/Sanya 266100/572024, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao 266003, China
| | - Deyao Qian
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao/Sanya 266100/572024, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao 266003, China
| | - Xinlei Meng
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao/Sanya 266100/572024, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao 266003, China
| | - Liangmin Yu
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao/Sanya 266100/572024, China
- Sanya Oceanographic Institution, Sanya 572024, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao 266003, China
| | - Xuefeng Yan
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao/Sanya 266100/572024, China
- Sanya Oceanographic Institution, Sanya 572024, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao 266003, China
| | - Zhiyu He
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao/Sanya 266100/572024, China
- Sanya Oceanographic Institution, Sanya 572024, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao 266003, China
| |
Collapse
|
15
|
Wang Y, Heymann F, Peiseler M. Intravital imaging: dynamic insights into liver immunity in health and disease. Gut 2024; 73:1364-1375. [PMID: 38777574 DOI: 10.1136/gutjnl-2023-331739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024]
Abstract
Inflammation is a critical component of most acute and chronic liver diseases. The liver is a unique immunological organ with a dense vascular network, leading to intense crosstalk between tissue-resident immune cells, passenger leucocytes and parenchymal cells. During acute and chronic liver diseases, the multifaceted immune response is involved in disease promoting and repair mechanisms, while upholding core liver immune functions. In recent years, single-cell technologies have unravelled a previously unknown heterogeneity of immune cells, reshaping the complexity of the hepatic immune response. However, inflammation is a dynamic biological process, encompassing various immune cells, orchestrated in temporal and spatial dimensions, and driven by multiorgan signals. Intravital microscopy (IVM) has emerged as a powerful tool to investigate immunity by visualising the dynamic interplay between different immune cells and their surroundings within a near-natural environment. In this review, we summarise the experimental considerations to perform IVM and highlight recent technological developments. Furthermore, we outline the unique contributions of IVM to our understanding of liver immunity. Through the lens of liver disease, we discuss novel immune-mediated disease mechanisms uncovered by imaging-based studies.
Collapse
Affiliation(s)
- Yuting Wang
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Felix Heymann
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Moritz Peiseler
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health at Charité, Berlin, Germany
| |
Collapse
|
16
|
Rycyk-Bojarzynska A, Kasztelan-Szczerbinska B, Cichoz-Lach H, Surdacka A, Rolinski J. Neutrophil PAD4 Expression and Its Pivotal Role in Assessment of Alcohol-Related Liver Disease. Int J Mol Sci 2024; 25:7597. [PMID: 39062840 PMCID: PMC11276781 DOI: 10.3390/ijms25147597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/02/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Neutrophils release neutrophil extracellular traps (NETs) as a defense strategy in response to broad-spectrum infections and sterile triggers. NETs consist of a DNA scaffold decorated with antimicrobial peptides (AMPs) and enzymatically active proteases, including peptidyl arginine deiminase type 4 (PAD4). Susceptibility to infections and inflammatory dysregulation are hallmarks of alcohol-related liver disease (ALD). Sixty-two patients with ALD were prospectively recruited, and they were followed for 90 days. Twenty-four healthy volunteers served as the control group. PAD4 concentrations were quantified using immunoenzymatic ELISAs. Correlation coefficients between PAD4 blood concentrations and markers of systemic inflammation; liver dysfunction severity scores; and ALD complications were calculated. The receiver operating curves (ROCs) and their areas under the curve (AUCs) were checked in order to assess the accuracy of PAD4 expression in predicting the degree of liver failure and the development of ALD complications. Systemic concentrations of PAD4 were significantly increased in the patients with ALD in comparison with controls. PAD4 levels correlated with the standard markers of inflammation and revealed a good predictive AUC (0.76) for survival in the whole ALD group. PAD4 seems to be an inflammatory mediator and may be potentially applied as a predictor of patient survival in ALD.
Collapse
Affiliation(s)
- Anna Rycyk-Bojarzynska
- Department of Gastroenterology with Endoscopy Unit, Medical University of Lublin, 20-090 Lublin, Poland
| | | | - Halina Cichoz-Lach
- Department of Gastroenterology with Endoscopy Unit, Medical University of Lublin, 20-090 Lublin, Poland
| | - Agata Surdacka
- Department of Clinical Immunology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Jacek Rolinski
- Department of Clinical Immunology, Medical University of Lublin, 20-093 Lublin, Poland
| |
Collapse
|
17
|
Ogino N, Leite MF, Guerra MT, Kruglov E, Asashima H, Hafler DA, Ito T, Pereira JP, Peiffer BJ, Sun Z, Ehrlich BE, Nathanson MH. Neutrophils insert elastase into hepatocytes to regulate calcium signaling in alcohol-associated hepatitis. J Clin Invest 2024; 134:e171691. [PMID: 38916955 PMCID: PMC11324315 DOI: 10.1172/jci171691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/20/2024] [Indexed: 06/27/2024] Open
Abstract
Neutrophil infiltration occurs in a variety of liver diseases, but it is unclear how neutrophils and hepatocytes interact. Neutrophils generally use granule proteases to digest phagocytosed bacteria and foreign substances or neutralize them in neutrophil extracellular traps. In certain pathological states, granule proteases play a destructive role against the host as well. More recently, nondestructive actions of neutrophil granule proteins have been reported, such as modulation of tissue remodeling and metabolism. Here, we report a completely different mechanism by which neutrophils act nondestructively, by inserting granules directly into hepatocytes. Specifically, elastase-containing granules were transferred to hepatocytes where elastase selectively degraded intracellular calcium channels to reduce cell proliferation without cytotoxicity. In response, hepatocytes increased expression of Serpin E2 and A3, which inhibited elastase activity. Elastase insertion was seen in patient specimens of alcohol-associated hepatitis, and the relationship between elastase-mediated ITPR2 degradation and reduced cell proliferation was confirmed in mouse models. Moreover, neutrophils from patients with alcohol-associated hepatitis were more prone to degranulation and more potent in reducing calcium channel expression than neutrophils from healthy individuals. This nondestructive and reversible action on hepatocytes defines a previously unrecognized role for neutrophils in the transient regulation of epithelial calcium signaling mechanisms.
Collapse
Affiliation(s)
- Noriyoshi Ogino
- Yale Liver Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - M. Fatima Leite
- Yale Liver Center, Yale University School of Medicine, New Haven, Connecticut, USA
- INCT - NanoBiofar – Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Mateus T. Guerra
- Yale Liver Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Emma Kruglov
- Yale Liver Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | | | - Takeshi Ito
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - João P. Pereira
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Brandon J. Peiffer
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Zhaoli Sun
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Barbara E. Ehrlich
- Yale Liver Center, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Pathology, New York University School of Medicine, New York, New York, USA
| | - Michael H. Nathanson
- Yale Liver Center, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
18
|
Ruan J, Tian Q, Li S, Zhou X, Sun Q, Wang Y, Xiao Y, Li M, Chang K, Yi X. The IL-33-ST2 axis plays a vital role in endometriosis via promoting epithelial-mesenchymal transition by phosphorylating β-catenin. Cell Commun Signal 2024; 22:318. [PMID: 38858740 PMCID: PMC11163813 DOI: 10.1186/s12964-024-01683-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/27/2024] [Indexed: 06/12/2024] Open
Abstract
OBJECTIVES Interleukin 33 (IL-33) is a crucial inflammatory factor that functions as an alarm signal in endometriosis (EMs). Epithelial-mesenchymal transition (EMT), a process related to inflammatory signals, intracellular reactive oxygen species (ROS) production, and lipid peroxidation, have been proposed as potential mechanisms that contribute to the development and progression of EMs. IL-33 is highly upregulated in the ectopic milieu. Moreover, ectopic endometrial cells constitutively express interleukin-33 receptor ST2 (IL-33R). However, the role of IL-33/ST2 in the EMT of EMs remains largely unknown. In this study, we aimed to mechanistically determine the role of IL-33/ST2 in EMs-associated fibrosis. MATERIALS AND METHODS We established a non-lethal oxidative stress model to explore the conditions that trigger IL-33 induction. We performed α-smooth muscle actin (α-SMA) protein detection, cell counting kit-8 (CCK-8) assays, and scratch assays to analyze the impact of IL-33 on primary endometrial stromal cells (ESCs) proliferation and invasion. Clinical samples from patients with or without EMs were subjected to immunohistochemical (IHC) and and immunofluorescence(IF) staining to assess the clinical relevance of IL-33 receptor ST2 and EMT-related proteins. Furthermore, we used the ectopic human endometrial epithelial cell line 12Z and normal human epithelial cell line EEC to evaluate the effects of IL-33 on Wnt/β-catenin signaling. The effect of IL-33 on EMT-associated fibrosis was validated in vivo by intraperitoneal injections of IL-33 and antiST2. RESULTS We observed that ectopic milieu, characterized by ROS, TGF-β1, and high level of estrogen, triggers the secretion of IL-33 from ectopic ESCs. Ectopic endometrial lesions exhibited higher level of fibrotic characteristics and ST2 expression than that in the normal endometrium. Exogenous recombinant human (rhIL-33) enhanced ESC migration and survival. Similarly, 12Z cells displayed a higher degree of EMT characteristics with elevated expression of CCN4 and Fra-1, downstream target genes of the WNT/β-catenin pathway, than that observed in EECs. Conversely, blocking IL-33 with neutralizing antibodies, knocking down ST2 or β-catenin with siRNA, and β-catenin dephosphorylation abolished its effects on EMT promotion. In vivo validation demonstrated that IL-33 significantly promotes EMs-related fibrosis through the activation of Wnt/β-catenin signaling. CONCLUSION Our data strongly support the vital role of the IL-33/ST2 pathway in EMs-associated fibrosis and emphasize the importance of the EMT in the pathophysiology of fibrosis. Targeting the IL-33/ST2/Wnt/β-catenin axis may hold promise as a feasible therapeutic approach for controlling fibrosis in EMs.
Collapse
Affiliation(s)
- Jingyao Ruan
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China
| | - Qi Tian
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China
| | - Siting Li
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China
| | - Xiaoyu Zhou
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China
| | - Qianzhi Sun
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China
| | - Yuning Wang
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China
| | - Yinping Xiao
- Department of Pathology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Mingqing Li
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Kaikai Chang
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China.
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China.
| | - Xiaofang Yi
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China.
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China.
| |
Collapse
|
19
|
Hintermann E, Tondello C, Fuchs S, Bayer M, Pfeilschifter JM, Taubert R, Mollenhauer M, Elferink RPJO, Manns MP, Christen U. Blockade of neutrophil extracellular trap components ameliorates cholestatic liver disease in Mdr2 (Abcb4) knockout mice. J Autoimmun 2024; 146:103229. [PMID: 38653165 DOI: 10.1016/j.jaut.2024.103229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 03/21/2024] [Accepted: 04/09/2024] [Indexed: 04/25/2024]
Abstract
Primary sclerosing cholangitis (PSC) is an (auto)immune-mediated cholestatic liver disease with a yet unclear etiology. Increasing evidence points to an involvement of neutrophils in chronic liver inflammation and cirrhosis but also liver repair. Here, we investigate the role of the neutrophil extracellular trap (NET) component myeloperoxidase (MPO) and the therapeutic potential of DNase I and of neutrophil elastase (NE) inhibitor GW311616A on disease outcome in the multidrug resistance 2 knockout (Mdr2-/-) mouse, a PSC animal model. Initially, we observed the recruitment of MPO expressing cells and the formation of NETs in liver biopsies of PSC patients and in Mdr2-/- livers. Furthermore, sera of Mdr2-/- mice contained perinuclear anti-neutrophil cytoplasmic antibody (p-ANCA)-like reactivity similar to PSC patient sera. Also, hepatic NE activity was significantly higher in Mdr2-/- mice than in wild type littermates. Flow cytometry analyses revealed that during disease development a highly active neutrophil subpopulation established specifically in the liver of Mdr2-/- mice. However, absence of their MPO activity, as in MPO-deficient Mdr2-/- mice, showed no effect on hepatobiliary disease severity. In contrast, clearance of extracellular DNA by DNase I reduced the frequency of liver-resident neutrophils, plasmacytoid dendritic cells (pDCs) and CD103+ conventional DCs and decreased cholangiocyte injury. Combination of DNase I with a pDC-depleting antibody was additionally hepatocyte-protective. Most importantly, GW311616A, an orally bioavailable inhibitor of human NE, attenuated hepatobiliary injury in a TNFα-dependent manner and damped hyperproliferation of biliary epithelial cells. Further, hepatic immigration and activity of CD11b+ DCs as well as the secretion of IFNγ by hepatic CD4 and CD8 T cells were reduced. Our findings delineate neutrophils as important participants in the immune cell crosstalk that drives cholestatic liver disease and identify NET components as potential therapeutic targets.
Collapse
Affiliation(s)
- Edith Hintermann
- Pharmazentrum Frankfurt / ZAFES, Goethe University, University Hospital Frankfurt, Frankfurt am Main, Germany.
| | - Camilla Tondello
- Pharmazentrum Frankfurt / ZAFES, Goethe University, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Sina Fuchs
- Pharmazentrum Frankfurt / ZAFES, Goethe University, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Monika Bayer
- Pharmazentrum Frankfurt / ZAFES, Goethe University, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Josef M Pfeilschifter
- Pharmazentrum Frankfurt / ZAFES, Goethe University, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Richard Taubert
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Germany
| | - Martin Mollenhauer
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Roland P J Oude Elferink
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands; European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Germany
| | - Michael P Manns
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Germany
| | - Urs Christen
- Pharmazentrum Frankfurt / ZAFES, Goethe University, University Hospital Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
20
|
Xiao MH, Wu S, Liang P, Ma D, Zhang J, Chen H, Zhong Z, Liu J, Jiang H, Feng X, Luo Z. Mucosal-associated invariant T cells promote ductular reaction through amphiregulin in biliary atresia. EBioMedicine 2024; 103:105138. [PMID: 38678809 PMCID: PMC11077624 DOI: 10.1016/j.ebiom.2024.105138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 05/01/2024] Open
Abstract
BACKGROUND Biliary atresia (BA) is a neonatal fibro-inflammatory cholangiopathy with ductular reaction as a key pathogenic feature predicting poor survival. Mucosal-associated invariant T (MAIT) cells are enriched in human liver and display multiple roles in liver diseases. We aimed to investigate the function of MAIT cells in BA. METHODS First, we analyzed correlations between liver MAIT cell and clinical parameters (survival, alanine transaminase, bilirubin, histological inflammation and fibrosis) in two public cohorts of patients with BA (US and China). Kaplan-Meier survival analysis and spearman correlation analysis were employed for survival data and other clinical parameters, respectively. Next, we obtained liver samples or peripheral blood from BA and control patients for bulk RNA sequencing, flow cytometry analysis, immunostaning and functional experiments of MAIT cells. Finally, we established two in vitro co-culture systems, one is the rhesus rotavirus (RRV) infected co-culture system to model immune dysfunction of human BA which was validated by single cell RNA sequencing and the other is a multicellular system composed of biliary organoids, LX-2 and MAIT cells to evaluate the role of MAIT cells on ductular reaction. FINDINGS Liver MAIT cells in BA were positively associated with low survival and ductular reaction. Moreover, liver MAIT cells were activated, exhibited a wound healing signature and highly expressed growth factor Amphiregulin (AREG) in a T cell receptor (TCR)-dependent manner. Antagonism of AREG abrogated the proliferative effect of BA MAIT cells on both cholangiocytes and biliary organoids. A RRV infected co-culture system, recapitulated immune dysfunction of human BA, disclosed that RRV-primed MAIT cells promoted cholangiocyte proliferation via AREG, and further induced inflammation and fibrosis in the multicellular system. INTERPRETATION MAIT cells exhibit a wound healing signature depending on TCR signaling and promote ductular reaction via AREG, which is associated with advanced fibrosis and predictive of low survival in BA. FUNDING This work was funded by National Natural Science Foundation of China grant (82001589 and 92168108), National Key R&D Program of China (2023YFA1801600) and by Basic and Applied Basic Research Foundation of Guangdong (2020A1515110921).
Collapse
Affiliation(s)
- Man-Huan Xiao
- Department of Pediatric Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Sihan Wu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Peishi Liang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Dong Ma
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Jiang Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Huadong Chen
- Department of Pediatric Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Zhihai Zhong
- Department of Pediatric Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Juncheng Liu
- Department of Pediatric Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Hong Jiang
- Department of Pediatric Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
| | - Xuyang Feng
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
| | - Zhenhua Luo
- Department of Pediatric Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
| |
Collapse
|
21
|
Mavila N, Siraganahalli Eshwaraiah M, Kennedy J. Ductular Reactions in Liver Injury, Regeneration, and Disease Progression-An Overview. Cells 2024; 13:579. [PMID: 38607018 PMCID: PMC11011399 DOI: 10.3390/cells13070579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/11/2024] [Accepted: 03/22/2024] [Indexed: 04/13/2024] Open
Abstract
Ductular reaction (DR) is a complex cellular response that occurs in the liver during chronic injuries. DR mainly consists of hyper-proliferative or reactive cholangiocytes and, to a lesser extent, de-differentiated hepatocytes and liver progenitors presenting a close spatial interaction with periportal mesenchyme and immune cells. The underlying pathology of DRs leads to extensive tissue remodeling in chronic liver diseases. DR initiates as a tissue-regeneration mechanism in the liver; however, its close association with progressive fibrosis and inflammation in many chronic liver diseases makes it a more complicated pathological response than a simple regenerative process. An in-depth understanding of the cellular physiology of DRs and their contribution to tissue repair, inflammation, and progressive fibrosis can help scientists develop cell-type specific targeted therapies to manage liver fibrosis and chronic liver diseases effectively.
Collapse
Affiliation(s)
- Nirmala Mavila
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (M.S.E.); (J.K.)
- Division of Applied Cell Biology and Physiology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Mallikarjuna Siraganahalli Eshwaraiah
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (M.S.E.); (J.K.)
| | - Jaquelene Kennedy
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (M.S.E.); (J.K.)
| |
Collapse
|
22
|
Hu Y, Bao X, Zhang Z, Chen L, Liang Y, Qu Y, Zhou Q, Zhou X, Fang J, Xiao Z, Fu Y, Yang H, Liu W, Lv Y, Cao H, Chen G, Ping J, Zhang H, Mu Y, Liu C, Lin CP, Wu J, Liu P, Chen J. Hepatic progenitor cell-originated ductular reaction facilitates liver fibrosis through activation of hedgehog signaling. Theranostics 2024; 14:2379-2395. [PMID: 38646644 PMCID: PMC11024850 DOI: 10.7150/thno.91572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 03/17/2024] [Indexed: 04/23/2024] Open
Abstract
Background: It is poorly understood what cellular types participate in ductular reaction (DR) and whether DR facilitates recovery from injury or accelerates hepatic fibrosis. The aim of this study is to gain insights into the role of hepatic progenitor cell (HPC)-originated DR during fibrotic progression. Methods: DR in liver specimens of PBC, chronic HBV infection (CHB) or NAFLD, and four rodent fibrotic models by different pathogenic processes was evaluated. Gli1 expression was inhibited in rodent models or cell culture and organoid models by AAV-shGli1 or treating with GANT61. Results: Severity of liver fibrosis was positively correlated with DR extent in patients with PBC, CHB or NAFLD. HPCs were activated, expanded, differentiated into reactive cholangiocytes and constituted "HPC-originated DR", accompanying with exacerbated fibrosis in rodent models of HPC activation & proliferation (CCl4/2-AAF-treated), Μdr2-/- spontaneous PSC, BDL-cholestatic fibrosis or WD-fed/CCl4-treated NASH-fibrosis. Gli1 expression was significantly increased in enriched pathways in vivo and in vitro. Enhanced Gli1 expression was identified in KRT19+-reactive cholangiocytes. Suppressing Gli1 expression by administration of AAV-shGli1 or GANT61 ameliorated HPC-originated DR and fibrotic extent. KRT19 expression was reduced after GANT61 treatment in sodium butyrate-stimulated WB-F344 cells or organoids or in cells transduced with Gli1 knockdown lentiviral vectors. In contrast, KRT19 expression was elevated after transducing Gli1 overexpression lentiviral vectors in these cells. Conclusions: During various modes of chronic injury, Gli1 acted as an important mediator of HPC activation, expansion, differentiation into reactive cholangiocytes that formed DR, and subsequently provoked hepatic fibrogenesis.
Collapse
Affiliation(s)
- Yonghong Hu
- Institute of Liver diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, China
- Institute of Surgery of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xinyu Bao
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
- Shanghai Clinical Research and Trial Center, Shanghai 201210, China
| | - Zheng Zhang
- Institute of Liver diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, China
| | - Long Chen
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yue Liang
- Institute of Liver diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, China
| | - Yan Qu
- Department of Hepatobiliary Surgery, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qun Zhou
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiaoxi Zhou
- Institute of Liver diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, China
| | - Jing Fang
- Institute of Liver diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, China
| | - Zhun Xiao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yadong Fu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hailin Yang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wei Liu
- Institute of Liver diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, China
| | - Ying Lv
- Institute of Liver diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, China
| | - Hongyan Cao
- Department of Gastroenterology, Shanghai University of Traditional Chinese Medicine Shanghai TCM - Integrated hospital, Shanghai 201203, China
| | - Gaofeng Chen
- Institute of Liver diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, China
| | - Jian Ping
- Institute of Liver diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, China
| | - Hua Zhang
- Institute of Liver diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, China
| | - Yongping Mu
- Institute of Liver diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, China
| | - Chenghai Liu
- Institute of Liver diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, China
| | - Chao-Po Lin
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
- Shanghai Clinical Research and Trial Center, Shanghai 201210, China
| | - Jian Wu
- Department of Medical Microbiology & Parasitology, MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai 200032, China
- Department of Gastroenterology & Hepatology, Zhongshan Hospital of Fudan University, Shanghai 200032, China
- Shanghai Institute of Liver Diseases, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Ping Liu
- Institute of Liver diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, China
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiamei Chen
- Institute of Liver diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, China
| |
Collapse
|
23
|
Wang J, Wang X, Peng H, Dong Z, Liangpunsakul S, Zuo L, Wang H. Platelets in Alcohol-Associated Liver Disease: Interaction With Neutrophils. Cell Mol Gastroenterol Hepatol 2024; 18:41-52. [PMID: 38461963 PMCID: PMC11127035 DOI: 10.1016/j.jcmgh.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 03/12/2024]
Abstract
Alcohol-associated liver disease (ALD) is a major contributor to liver-related mortality globally. An increasing body of evidence underscores the pivotal role of platelets throughout the spectrum of liver injury and recovery, offering unique insights into liver homeostasis and pathobiology. Alcoholic-associated steatohepatitis is characterized by the infiltration of hepatic neutrophils. Recent studies have highlighted the extensive distance neutrophils travel through sinusoids to reach the liver injury site, relying on a platelet-paved endothelium for efficient crawling. The adherence of platelets to neutrophils is crucial for accurate migration from circulation to the inflammatory site. A gradual decline in platelet levels leads to diminished neutrophil recruitment. Platelets exhibit the ability to activate neutrophils. Platelet activation is heightened upon the release of platelet granule contents, which synergistically activate neutrophils through their respective receptors. The sequence culminates in the formation of platelet-neutrophil complexes and the release of neutrophil extracellular traps intensifies liver damage, fosters inflammatory immune responses, and triggers hepatotoxic processes. Neutrophil infiltration is a hallmark of alcohol-associated steatohepatitis, and the roles of neutrophils in ALD pathogenesis have been studied extensively, however, the involvement of platelets in ALD has received little attention. The current review consolidates recent findings on the intricate and diverse roles of platelets and neutrophils in liver pathophysiology and in ALD. Potential therapeutic strategies are highlighted, focusing on targeting platelet-neutrophil interactions and activation in ALD. The anticipation is that innovative methods for manipulating platelet and neutrophil functions will open promising avenues for future ALD therapy.
Collapse
Affiliation(s)
- Juan Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, Anhui, China
| | - Xianda Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, Anhui, China
| | - Haodong Peng
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China; The First School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Zijian Dong
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, Anhui, China
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Li Zuo
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China; Laboratory of Molecular Biology, Department of Biochemistry, Anhui Medical University, Hefei, Anhui, China.
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
24
|
Wu Z, Liu D, Ou Y, Xu Z, Heng G, Liu W, Fu N, Wang J, Jiang D, Gan L, Dong J, Wang X, Chen Z, Zhang L, Zhang C. Mechanism and endoscopic-treatment-induced evolution of biliary non-anastomotic stricture after liver transplantation revealed by single-cell RNA sequencing. Clin Transl Med 2024; 14:e1622. [PMID: 38481381 PMCID: PMC10938070 DOI: 10.1002/ctm2.1622] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Biliary complications, especially non-anastomotic stricture (NAS), are the main complications after liver transplantation. Insufficient sampling and no recognized animal models obstruct the investigation. Thus, the mechanisms and alterations that occur during endoscopic treatment (ET) of NAS remain unclear. METHODS Samples were obtained with endoscopic forceps from the hilar bile ducts of NAS patients receiving continuous biliary stent implantation after diagnosis. Retrospective analysis of multiple studies indicated that the duration of ET for NAS was approximately 1-2 years. Thus, we divided the patients into short-term treatment (STT) and long-term treatment (LTT) groups based on durations of less or more than 1 year. Samples were subjected to single-cell RNA sequencing. Transcriptomic differences between STT and normal groups were defined as the NAS mechanism. Similarly, alterations from STT to LTT groups were regarded as endoscopic-treatment-induced evolution. RESULTS In NAS, inflammation and immune-related pathways were upregulated in different cell types, with nonimmune cells showing hypoxia pathway upregulation and immune cells showing ATP metabolism pathway upregulation, indicating heterogeneity. We confirmed a reduction in bile acid metabolism-related SPP1+ epithelial cells in NAS. Increases in proinflammatory and profibrotic fibroblast subclusters indicated fibrotic progression in NAS. Furthermore, immune disorders in NAS were exacerbated by an increase in plasma cells and dysfunction of NK and NKT cells. ET downregulated multicellular immune and inflammatory responses and restored epithelial and endothelial cell proportions. CONCLUSIONS This study reveals the pathophysiological and genetic mechanisms and evolution of NAS induced by ET, thereby providing preventive and therapeutic insights into NAS. HIGHLIGHTS For the first time, single-cell transcriptome sequencing was performed on the bile ducts of patients with biliary complications. scRNA-seq analysis revealed distinct changes in the proportion and phenotype of multiple cell types during Nonanastomotic stricture (NAS) and endoscopic treatment. A reduction in bile acid metabolism-related SPP1+ epithelial cells and VEGFA+ endothelial cells, along with explosive infiltration of plasma cells and dysfunction of T and NK cells in NAS patients. SPP1+ macrophages and BST2+ T cells might serve as a surrogate marker for predicting endoscopic treatment.
Collapse
Affiliation(s)
- Zhaoyi Wu
- Department of Hepatobiliary SurgerySouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingChina
| | - Danqing Liu
- Department of Hepatobiliary SurgerySouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingChina
| | - Yanjiao Ou
- Department of Hepatobiliary SurgerySouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingChina
| | - Zeliang Xu
- Department of Hepatobiliary SurgerySouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingChina
| | - Gang Heng
- Department of General SurgeryPLA Middle Military Command General HospitalWuhanChina
| | - Wei Liu
- Department of Hepatobiliary SurgerySouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingChina
| | - Nengsheng Fu
- Department of Hepatobiliary SurgerySouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingChina
| | - Jingyi Wang
- Department of Hepatobiliary SurgerySouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingChina
| | - Di Jiang
- Department of Hepatobiliary SurgerySouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingChina
| | - Lang Gan
- Department of Hepatobiliary SurgerySouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingChina
| | - Jiahong Dong
- Hepatopancereatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical MedicineTsinghua UniversityBeijingPeople's Republic of China
| | - Xiaojun Wang
- Department of Hepatobiliary SurgerySouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingChina
| | - Zhiyu Chen
- Department of Hepatobiliary SurgerySouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingChina
| | - Leida Zhang
- Department of Hepatobiliary SurgerySouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingChina
| | - Chengcheng Zhang
- Department of Hepatobiliary SurgerySouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingChina
| |
Collapse
|
25
|
Meng Q, Zhu H, Li Y, Peng X, Wang T, Huang H, Zhou H, Liu Y, Ru S, Wu J, Ma Y. Quantitative proteomics reveals the protective effects of Yinchenzhufu decoction against cholestatic liver fibrosis in mice by inhibiting the PDGFRβ/PI3K/AKT pathway. Front Pharmacol 2024; 15:1341020. [PMID: 38469403 PMCID: PMC10926276 DOI: 10.3389/fphar.2024.1341020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 01/25/2024] [Indexed: 03/13/2024] Open
Abstract
Introduction: Yinchenzhufu decoction (YCZFD) is a traditional Chinese medicine formula with hepatoprotective effects. In this study, the protective effects of YCZFD against cholestatic liver fibrosis (CLF) and its underlying mechanisms were evaluated. Methods: A 3, 5-diethoxycarbonyl-1, 4-dihydro-collidine (DDC)-induced cholestatic mouse model was used to investigate the amelioration of YCZFD on CLF. Data-independent acquisition-based mass spectrometry was performed to investigate proteomic changes in the livers of mice in three groups: control, model, and model treated with high-dose YCZFD. The effects of YCZFD on the expression of key proteins were confirmed in mice and cell models. Results: YCZFD significantly decreased the levels of serum biochemical, liver injury, and fibrosis indicators of cholestatic mice. The proteomics indicated that 460 differentially expressed proteins (DEPs) were identified among control, model, and model treated with high-dose YCZFD groups. Enrichment analyses of these DEPs revealed that YCZFD influenced multiple pathways, including PI3K-Akt, focal adhesion, ECM-receptor interaction, glutathione metabolism, and steroid biosynthesis pathways. The expression of platelet derived growth factor receptor beta (PDGFRβ), a receptor associated with the PI3K/AKT and focal adhesion pathways, was upregulated in the livers of cholestatic mice but downregulated by YCZFD. The effects of YCZFD on the expression of key proteins in the PDGFRβ/PI3K/AKT pathway were further confirmed in mice and transforming growth factor-β-induced hepatic stellate cells. We uncovered seven plant metabolites (chlorogenic acid, scoparone, isoliquiritigenin, glycyrrhetinic acid, formononetin, atractylenolide I, and benzoylaconitine) of YCZFD that may regulate PDGFRβ expression. Conclusion: YCZFD substantially protects against DDC-induced CLF mainly through regulating the PDGFRβ/PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Qian Meng
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Analytical Research Center for Organic and Biological Molecules, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Hongwen Zhu
- Analytical Research Center for Organic and Biological Molecules, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yuanyuan Li
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaotian Peng
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tianming Wang
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hui Huang
- Analytical Research Center for Organic and Biological Molecules, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Hu Zhou
- Analytical Research Center for Organic and Biological Molecules, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yuejia Liu
- Analytical Research Center for Organic and Biological Molecules, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Sujie Ru
- Analytical Research Center for Organic and Biological Molecules, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Jiasheng Wu
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yueming Ma
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
26
|
Abstract
Alcohol-associated liver disease (ALD) is a major cause of chronic liver disease worldwide, and comprises a spectrum of several different disorders, including simple steatosis, steatohepatitis, cirrhosis, and superimposed hepatocellular carcinoma. Although tremendous progress has been made in the field of ALD over the last 20 years, the pathogenesis of ALD remains obscure, and there are currently no FDA-approved drugs for the treatment of ALD. In this Review, we discuss new insights into the pathogenesis and therapeutic targets of ALD, utilizing the study of multiomics and other cutting-edge approaches. The potential translation of these studies into clinical practice and therapy is deliberated. We also discuss preclinical models of ALD, interplay of ALD and metabolic dysfunction, alcohol-associated liver cancer, the heterogeneity of ALD, and some potential translational research prospects for ALD.
Collapse
|
27
|
Krishnan A, Ozturk NB, Cutshaw KA, Guicciardi ME, Kitagataya T, Olson KE, Pavelko KD, Sherman W, Wixom AQ, Jalan-Sakrikar N, Baez-Faria M, Gutierrez F, Gores GJ. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) deletion in myeloid cells augments cholestatic liver injury. Sci Rep 2024; 14:2145. [PMID: 38273071 PMCID: PMC10810846 DOI: 10.1038/s41598-024-52710-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 01/23/2024] [Indexed: 01/27/2024] Open
Abstract
Ductular reactive (DR) cells exacerbate cholestatic liver injury and fibrosis. Herein, we posit that tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) emanates from recruited macrophages and restrains DR cell expansion, thereby limiting cholestatic liver injury. Wild type (WT), Trailfl/fl and myeloid-specific Trail deleted (TrailΔmye) C57BL/6 mice were exposed to DDC diet-induced cholestatic liver injury, which induced hepatomegaly and liver injury as compared to control diet-fed mice. However, parameters of liver injury, fibrosis, and inflammation were all increased in the TrailΔmye mice as compared to the WT and Trailfl/fl mice. High dimensional mass cytometry indicated that cholestasis resulted in increased hepatic recruitment of subsets of macrophages and neutrophils in the TrailΔmye mice. Spatial transcriptomics analysis revealed that the PanCK+ cholangiocytes from TrailΔmye mice had increased expression of the known myeloid attractants S100a8, Cxcl5, Cx3cl1, and Cxcl1. Additionally, in situ hybridization of Cxcl1, a potent neutrophil chemoattractant, demonstrated an increased expression in CK19+ cholangiocytes of TrailΔmye mice. Collectively, these data suggest that TRAIL from myeloid cells, particularly macrophages, restrains a subset of DR cells (i.e., Cxcl1 positive cells), limiting liver inflammation and fibrosis. Reprogramming macrophages to express TRAIL may be salutary in cholestasis.
Collapse
Affiliation(s)
- Anuradha Krishnan
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Nazli Begum Ozturk
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Kaiyel A Cutshaw
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Maria Eugenia Guicciardi
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Takashi Kitagataya
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Kirsta E Olson
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | | | - William Sherman
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Alexander Q Wixom
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Nidhi Jalan-Sakrikar
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Michelle Baez-Faria
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Florencia Gutierrez
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Gregory J Gores
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
28
|
Greenman R, Snir T, Katav A, Aricha R, Mishalian I, Hay O, Frankel M, Lawler J, Saffioti F, Pinzani M, Thorburn D, Peled A, Mor A, Vaknin I. The Role of CCL24 in Primary Sclerosing Cholangitis: Bridging Patient Serum Proteomics to Preclinical Data. Cells 2024; 13:209. [PMID: 38334601 PMCID: PMC10854794 DOI: 10.3390/cells13030209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/15/2024] [Accepted: 01/19/2024] [Indexed: 02/10/2024] Open
Abstract
Primary sclerosing cholangitis (PSC) is an inflammatory and fibrotic biliary disease lacking approved treatment. We studied CCL24, a chemokine shown to be overexpressed in damaged bile ducts, and its involvement in key disease-related mechanisms. Serum proteomics of PSC patients and healthy controls (HC) were analyzed using the Olink® proximity extension assay and compared based on disease presence, fibrosis severity, and CCL24 levels. Disease-related canonical pathways, upstream regulators, and toxicity functions were elevated in PSC patients compared to HC and further elevated in patients with high CCL24 levels. In vitro, a protein signature in CCL24-treated hepatic stellate cells (HSCs) differentiated patients by disease severity. In mice, CCL24 intraperitoneal injection selectively recruited neutrophils and monocytes. Treatment with CM-101, a CCL24-neutralizing antibody, in an α-naphthylisothiocyanate (ANIT)-induced cholestasis mouse model effectively inhibited accumulation of peribiliary neutrophils and macrophages while reducing biliary hyperplasia and fibrosis. Furthermore, in PSC patients, CCL24 levels were correlated with upregulation of monocyte and neutrophil chemotaxis pathways. Collectively, these findings highlight the distinct role of CCL24 in PSC, influencing disease-related mechanisms, affecting immune cells trafficking and HSC activation. Its blockade with CM-101 reduces inflammation and fibrosis and positions CCL24 as a promising therapeutic target in PSC.
Collapse
Affiliation(s)
| | - Tom Snir
- Chemomab Therapeutics Ltd., Tel Aviv 6158002, Israel
| | - Avi Katav
- Chemomab Therapeutics Ltd., Tel Aviv 6158002, Israel
| | | | - Inbal Mishalian
- Goldyne Savad Institute of Gene Therapy, Hebrew University Hospital Jerusalem, Jerusalem 91120, Israel
| | - Ophir Hay
- Goldyne Savad Institute of Gene Therapy, Hebrew University Hospital Jerusalem, Jerusalem 91120, Israel
| | | | - John Lawler
- Chemomab Therapeutics Ltd., Tel Aviv 6158002, Israel
| | - Francesca Saffioti
- UCL Institute for Liver and Digestive Health, University College of London, London NW3 2PF, UK
- Sheila Sherlock Liver Centre, Royal Free London NHS Foundation Trust, London NW3 2QG, UK
- Department of Gastroenterology and Hepatology, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Massimo Pinzani
- UCL Institute for Liver and Digestive Health, University College of London, London NW3 2PF, UK
- Sheila Sherlock Liver Centre, Royal Free London NHS Foundation Trust, London NW3 2QG, UK
| | - Douglas Thorburn
- UCL Institute for Liver and Digestive Health, University College of London, London NW3 2PF, UK
- Sheila Sherlock Liver Centre, Royal Free London NHS Foundation Trust, London NW3 2QG, UK
| | - Amnon Peled
- Goldyne Savad Institute of Gene Therapy, Hebrew University Hospital Jerusalem, Jerusalem 91120, Israel
| | - Adi Mor
- Chemomab Therapeutics Ltd., Tel Aviv 6158002, Israel
| | - Ilan Vaknin
- Chemomab Therapeutics Ltd., Tel Aviv 6158002, Israel
| |
Collapse
|
29
|
Ceci L, Gaudio E, Kennedy L. Cellular Interactions and Crosstalk Facilitating Biliary Fibrosis in Cholestasis. Cell Mol Gastroenterol Hepatol 2024; 17:553-565. [PMID: 38216052 PMCID: PMC10883986 DOI: 10.1016/j.jcmgh.2024.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 01/14/2024]
Abstract
Biliary fibrosis is seen in cholangiopathies, including primary biliary cholangitis (PBC) and primary sclerosing cholangitis (PSC). In PBC and PSC, biliary fibrosis is associated with worse outcomes and histologic scores. Within the liver, both hepatic stellate cells (HSCs) and portal fibroblasts (PFs) contribute to biliary fibrosis, but their roles can differ. PFs reside near the bile ducts and may be the first responders to biliary damage, whereas HSCs may be recruited later and initiate bridging fibrosis. Indeed, different models of biliary fibrosis can activate PFs and HSCs to varying degrees. The portal niche can be composed of cholangiocytes, HSCs, PFs, endothelial cells, and various immune cells, and interactions between these cell types drive biliary fibrosis. In this review, we discuss the mechanisms of biliary fibrosis and the roles of PFs and HSCs in this process. We will also evaluate cellular interactions and mechanisms that contribute to biliary fibrosis in different models and highlight future perspectives and potential therapeutics.
Collapse
Affiliation(s)
- Ludovica Ceci
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Sapienza, University of Rome, Italy
| | - Eugenio Gaudio
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Sapienza, University of Rome, Italy
| | - Lindsey Kennedy
- Department of Research, Richard L. Roudebush VA Medical Center, Indianapolis, Indiana; Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana.
| |
Collapse
|