1
|
Brummer C, Singer K, Renner K, Bruss C, Hellerbrand C, Dorn C, Reichelt-Wurm S, Gronwald W, Pukrop T, Herr W, Banas M, Kreutz M. The spleen-liver axis supports obesity-induced systemic and fatty liver inflammation via MDSC and NKT cell enrichment. Mol Cell Endocrinol 2025; 601:112518. [PMID: 40054835 DOI: 10.1016/j.mce.2025.112518] [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: 07/16/2024] [Revised: 01/29/2025] [Accepted: 03/05/2025] [Indexed: 04/01/2025]
Abstract
Obesity promotes adipose tissue inflammation and leads to impaired local but also systemic immune cell homeostasis. This chronic low-grade inflammation plays a significant role in the development of obesity-associated secondary diseases such as metabolic associated fatty liver disease or cancer. The spleen as the central organ of immune cell regulation is anatomically directly connected to the visceral adipose tissue and the liver via the portal vein circulation. However, the inter-organ crosstalk and linkage between obesity-induced systemic, hepatic and splenic immune cell dysregulation is not clearly outlined. In this study blood, spleen, and liver immune cells of non-obese wildtype vs. leptin deficient obese BTBR mice were isolated and analyzed in terms of leukocyte composition by flow cytometry. Significant differences between circulating, spleen- and liver-resident immune cell distribution revealed, that obesity-induced hepatic and systemic immune cell dysregulation is distinct from splenic immune cell reprogramming. Fatty liver inflammation was associated with splenic myeloid derived suppressor cell (MDSC) and natural killer T cell (NKT) enrichment whereas loss of hepatic T and B cells was not reflected by the splenic lymphocyte landscape. Correlation analysis confirmed a selective strong positive correlation between spleen and liver MDSC and NKT cell distribution indicating that the spleen-liver axis modulates obesity-induced immune dysregulation in a cell-specific manner. Similar results were observed in a diet-induced obesity mouse model. These data provide novel insights into the role of the spleen-liver axis in obesity-induced inflammation and foster the understanding of obesity-associated complications such as fatty liver disease and cancer.
Collapse
Affiliation(s)
- Christina Brummer
- Department of Internal Medicine III, University Hospital Regensburg, 93053, Regensburg, Germany.
| | - Katrin Singer
- Department of Internal Medicine III, University Hospital Regensburg, 93053, Regensburg, Germany
| | - Kathrin Renner
- Department of Otorhinolaryngology, University Hospital Regensburg, 93053, Regensburg, Germany
| | - Christina Bruss
- Department of Internal Medicine III, University Hospital Regensburg, 93053, Regensburg, Germany; Department of Gynecology and Obstetrics, University Hospital Regensburg, 93053, Regensburg, Germany
| | - Claus Hellerbrand
- Institute of Biochemistry, University of Erlangen, 91054, Erlangen, Germany
| | - Christoph Dorn
- Institute of Pharmacy, University of Regensburg, 93053, Regensburg, Germany
| | - Simone Reichelt-Wurm
- Department of Nephrology, University Hospital Regensburg, 93053, Regensburg, Germany
| | - Wolfram Gronwald
- Institute of Functional Genomics, University of Regensburg, 93053, Regensburg, Germany
| | - Tobias Pukrop
- Department of Internal Medicine III, University Hospital Regensburg, 93053, Regensburg, Germany; Comprehensive Cancer Center Ostbayern (CCCO), 93053, Regensburg, Germany
| | - Wolfgang Herr
- Department of Internal Medicine III, University Hospital Regensburg, 93053, Regensburg, Germany
| | - Miriam Banas
- Department of Nephrology, University Hospital Regensburg, 93053, Regensburg, Germany
| | - Marina Kreutz
- Department of Internal Medicine III, University Hospital Regensburg, 93053, Regensburg, Germany
| |
Collapse
|
2
|
Kuchay MS, Choudhary NS, Ramos-Molina B. Pathophysiological underpinnings of metabolic dysfunction-associated steatotic liver disease. Am J Physiol Cell Physiol 2025; 328:C1637-C1666. [PMID: 40244183 DOI: 10.1152/ajpcell.00951.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 01/31/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is emerging as the leading cause of chronic liver disease worldwide, reflecting the global epidemics of obesity, metabolic syndrome, and type 2 diabetes. Beyond its strong association with excess adiposity, MASLD encompasses a heterogeneous population that includes individuals with normal body weight ("lean MASLD") highlighting the complexity of its pathogenesis. This disease results from a complex interplay between genetic susceptibility, epigenetic modifications, and environmental factors, which converge to disrupt metabolic homeostasis. Adipose tissue dysfunction and insulin resistance trigger an overflow of lipids to the liver, leading to mitochondrial dysfunction, oxidative stress, and hepatocellular injury. These processes promote hepatic inflammation and fibrogenesis, driven by cross talk among hepatocytes, immune cells, and hepatic stellate cells, with key contributions from gut-liver axis perturbations. Recent advances have unraveled pivotal molecular pathways, such as transforming growth factor-β signaling, Notch-induced osteopontin, and sphingosine kinase 1-mediated responses, that orchestrate fibrogenic activation. Understanding these interconnected mechanisms is crucial for developing targeted therapies. This review integrates current knowledge on the pathophysiology of MASLD, emphasizing emerging concepts such as lean metabolic dysfunction-associated steatohepatitis (MASH), epigenetic alterations, hepatic extracellular vesicles, and the relevance of extrahepatic signals. It also discusses novel therapeutic strategies under investigation, aiming to provide a comprehensive and structured overview of the evolving MASLD landscape for both basic scientists and clinicians.
Collapse
Affiliation(s)
| | - Narendra Singh Choudhary
- Institute of Digestive and Hepatobiliary Sciences, Medanta-The Medicity Hospital, Gurugram, India
| | - Bruno Ramos-Molina
- Group of Obesity, Diabetes & Metabolism, Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| |
Collapse
|
3
|
Wu Q, Yang Y, Lin S, Geller DA, Yan Y. The microenvironment in the development of MASLD-MASH-HCC and associated therapeutic in MASH-HCC. Front Immunol 2025; 16:1569915. [PMID: 40370443 PMCID: PMC12074932 DOI: 10.3389/fimmu.2025.1569915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 04/08/2025] [Indexed: 05/16/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a series of obesity-related metabolic liver diseases, ranging from relatively benign hepatic steatosis to metabolic-associated steatohepatitis (MASH). With the changes in lifestyle, its incidence and prevalence have risen to epidemic proportions globally. In recent years, an increasing amount of evidence has indicated that the hepatic microenvironment is involved in the pathophysiological processes of MASH-induced liver fibrosis and the formation of hepatocellular carcinoma (HCC). The hepatic microenvironment is composed of various parenchymal and non-parenchymal cells, which communicate with each other through various factors. In this review, we focus on the changes in hepatocytes, cholangiocytes, liver sinusoidal endothelial cells (LSECs), hepatic stellate cells (HSCs), Kupffer cells (KC), dendritic cells (DC), neutrophils, monocytes, T and B lymphocytes, natural killer cells (NK), natural killer T cells (NKT), mucosal-associated invariant T cells (MAIT), γδT cells, and gut microbiota during the progression of MASLD. Furthermore, we discuss promising therapeutic strategies targeting the microenvironment of MASLD-MASH-HCC.
Collapse
Affiliation(s)
- Qiulin Wu
- Department of General Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yan Yang
- Department of General Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Shixun Lin
- Department of General Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - David A. Geller
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Yihe Yan
- Department of General Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
4
|
Yan X, Li J, Wang Q, Xu T, Wu D. Peripheral blood lymphocyte subsets of Chinese adults with liver steatosis. Sci Rep 2025; 15:14970. [PMID: 40301513 PMCID: PMC12041267 DOI: 10.1038/s41598-025-99510-x] [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: 07/05/2024] [Accepted: 04/21/2025] [Indexed: 05/01/2025] Open
Abstract
To evaluate the effect of liver steatosis on systematic cellular immunity through the changes of peripheral blood lymphocyte subsets, we retrospectively reviewed subjects receiving lymphocyte subtyping during annual medical check-ups. Liver steatosis was detected by abdominal computerized tomography or ultrasound. Immunophenotyping of peripheral blood lymphocytes was analyzed by flow cytometry. Cell counts of lymphocyte subsets were calculated using a dual-platform method. The relationship between lymphocyte subsets and liver steatosis was analyzed using multivariate linear models. Using the database from January 2017 to December 2022, we included 5042 subjects, including 1441 participants with liver steatosis. After adjusting for age, gender, and metabolic dysfunctions, the presence of liver steatosis increased the absolute values of CD19+, CD16+56+, CD3+, CD4+, CD4+CD28+, CD4+CD45RA-, CD4+CD45RA+, CD4+CD45RA+62L+, CD8+, CD8+CD28+, CD8+DR+, and CD8+CD38+ cell. In males, T lymphocyte counts of all T subsets increased in liver steatosis. In females, the significant differences in subsets included increased CD4+CD28+, CD4+CD45RA-, CD8+, and CD8+DR+ cells. Significant decreases were revealed in functional and naïve T cells with aging. Metabolic factors such as hypertension and abnormal glucose metabolism increase CD4+, CD4+CD28+, and CD4+CD45RA subsets. Hyperlipidaemia appears not to affect T cell counts, whereas obesity has some effect on both CD4+ (β = 0.041, 95% CI 0.032-0.051) and CD8+ cells (β = 0.036, 95% CI 0.025-0.048). Liver steatosis potentially affects peripheral blood lymphocyte subsets, while age, gender, and metabolic dysfunctions are also associated with these immune alterations.
Collapse
Affiliation(s)
- Xiaxiao Yan
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Li
- Department of Health Management, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Qiang Wang
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tengda Xu
- Department of Health Management, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Dong Wu
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
5
|
Rabiu L, Zhang P, Liu Z, Tang Y, Gidado KI, Ibrahim A, Saliu MA, Tariq HK, Wan X, Xu S, Xu Z, Zhang G. TIPE2 deficiency amplifies inflammation and immune dysregulation in MASH through modulating hepatic lipid metabolism and immune cell function. Inflamm Res 2025; 74:65. [PMID: 40244311 DOI: 10.1007/s00011-025-02031-6] [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: 12/24/2024] [Revised: 03/20/2025] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Metabolic Dysfunction-Associated Steatohepatitis (MASH) affects nearly 25% of the global population, yet there are no effective pharmacological treatments. Tumor necrosis factor α-induced protein 8-like 2 (TIPE2) is expressed in various immune cells and is crucial for regulating both innate and adaptive immune responses. However, its role in MASH development and the underlying mechanisms remain unclear. METHOD In this study, the role of TIPE2 in MASH was investigated using TIPE2 knockout (KO) mice and human hepatic LO2 cells. Immune cell infiltration, cytokine levels, and gene expression were analyzed. Techniques included flow cytometry for immune cell profiling, cytokine analysis, RNA sequencing, and quantitative PCR (qPCR) for validating gene expression changes. RESULTS TIPE2 was identified as a key regulator in MASH, influencing immune modulation and metabolic processes. TIPE2 KO mice exhibited increased infiltration and activation of natural killer (NK) cells, M1 macrophages, and myeloid-derived suppressor cells (MDSCs), along with elevated pro-inflammatory cytokines such as IFN-gamma, TNF-alpha, IL- 1 beta, and IL- 6. MDSCs from TIPE2 KO mice demonstrated enhanced PD-L1 expression, contributing to chronic liver inflammation through T cell suppression. RNA sequencing revealed that TIPE2 overexpression in human hepatic LO2 cells upregulated genes associated with amino acid biosynthesis, carbon metabolism, lipid regulation, glycolysis, and gluconeogenesis. These findings were supported by qPCR analyses of liver samples from mice, confirming TIPE2's role in maintaining lipid homeostasis and modulating immune responses. CONCLUSION The study highlights the pivotal role of TIPE2 in immune regulation and its influence on immune cell activation and inflammatory responses, which are critical in MASH progression. By exploring TIPE2-mediated immune regulation and its impact on the interplay between immune cell dynamics and liver metabolism, this research underscores TIPE2's central role in linking immune dysfunction to metabolic disturbances in MASH.
Collapse
Affiliation(s)
- Lawan Rabiu
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Pengchao Zhang
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Zhongming Liu
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Yexiao Tang
- Cancer Center, Shenzhen Guangming District People's Hospital, Shenzhen, 518106, PR China
| | - Khalid I Gidado
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Abdulrahman Ibrahim
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Muhammad A Saliu
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Hafiza Kashaf Tariq
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Xiaochun Wan
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Shu Xu
- Cancer Center, Shenzhen Guangming District People's Hospital, Shenzhen, 518106, PR China.
| | - Zhiming Xu
- Cancer Center, Shenzhen Guangming District People's Hospital, Shenzhen, 518106, PR China.
| | - Guizhong Zhang
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China.
- University of Chinese Academy of Sciences, Beijing, 100049, PR China.
| |
Collapse
|
6
|
Vizioli G, Nicoletti A, Feliciani D, Funaro B, Zileri Dal Verme L, Ponziani FR, Zocco MA, Gasbarrini A, Gabrielli M. Immunotherapy and MASLD-Related HCC: Should We Reconsider the Role of Etiology in the Therapeutic Approach to HCC? APPLIED SCIENCES 2025; 15:2279. [DOI: 10.3390/app15052279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/21/2025]
Abstract
Hepatocellular carcinoma (HCC) accounts for 90% of primary liver cancers and typically arises in the context of chronic liver disease. With the increasing prevalence of metabolic disorders, metabolic dysfunction-associated steatotic liver disease (MASLD) has become the leading cause of chronic liver disease and the most rapidly increasing cause of HCC. The role of dysfunctional innate and adaptive immune responses in the development and progression of HCC is well-established, prompting numerous trials to evaluate the efficacy of immune checkpoint inhibitors (ICIs) in targeting tumor cells. These trials have yielded promising results, and ICIs, in combination with anti-vascular endothelial growth factor (VEGF) monoclonal antibodies, are now approved as first-line therapy for patients with metastatic or unresectable HCC, irrespective of the underlying liver disease. Notably, MASLD itself is characterized by immune system dysfunction, as metabolic inflammation plays a central role in its onset and progression. However, clinical studies and post-hoc analyses suggest that immunotherapy may be less effective in MASLD-associated HCC compared to viral-related HCC. This emerging evidence raises the question of whether the underlying liver disease influences the therapeutic response to ICIs in HCC. It may be time to consider tailoring therapeutic strategies for HCC based on the specific etiological, histological, and genotypical subgroups.
Collapse
Affiliation(s)
- Giuseppina Vizioli
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Alberto Nicoletti
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Daniela Feliciani
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Barbara Funaro
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Lorenzo Zileri Dal Verme
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Francesca Romana Ponziani
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Maria Assunta Zocco
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Maurizio Gabrielli
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
7
|
Burtis AE, DeNicola DM, Ferguson ME, Santos RG, Pinilla C, Kriss MS, Orlicky DJ, Tamburini BAJ, Gillen AE, Burchill MA. Ag-driven CD8 + T cell clonal expansion is a prominent feature of MASH in humans and mice. Hepatology 2025; 81:591-608. [PMID: 39047085 PMCID: PMC11737124 DOI: 10.1097/hep.0000000000000971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 05/31/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND AND AIMS Chronic liver disease due to metabolic dysfunction-associated steatohepatitis (MASH) is a rapidly increasing global epidemic. MASH progression is a consequence of the complex interplay between inflammatory insults and dysregulated hepatic immune responses. T lymphocytes have been shown to accumulate in the liver during MASH, but the cause and consequence of T cell accumulation in the liver remain unclear. Our study aimed to define the phenotype and T cell receptor diversity of T cells from human cirrhotic livers and an animal model of MASH to begin resolving their function in disease. APPROACH AND RESULTS In these studies, we evaluated differences in T cell phenotype in the context of liver disease. Accordingly, we isolated liver resident T cell populations from humans with cirrhosis and from mice with diet-induced MASH. Using both 5' single-cell sequencing and flow cytometry, we defined the phenotype and T cell receptor repertoire of liver resident T cells during health and disease. CONCLUSIONS MASH-induced human cirrhosis and diet-induced MASH in mice resulted in the accumulation of activated and clonally expanded T cells in the liver. The clonally expanded T cells in the liver expressed markers of chronic antigenic stimulation, including PD1 , TIGIT , and TOX . Overall, this study establishes for the first time that T cells undergo Ag-dependent clonal expansion and functional differentiation during the progression of MASH. These studies could lead to the identification of antigenic targets that drive T cell activation, clonal expansion, and recruitment to the liver during MASH.
Collapse
Affiliation(s)
- Abbigayl E.C. Burtis
- Division of Gastroenterology and Hepatology, Department of Medicine, Aurora, Colorado, USA
- Molecular Biology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Destiny M.C. DeNicola
- Division of Gastroenterology and Hepatology, Department of Medicine, Aurora, Colorado, USA
- Molecular Biology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Megan E. Ferguson
- Division of Gastroenterology and Hepatology, Department of Medicine, Aurora, Colorado, USA
| | - Radleigh G. Santos
- Department of Mathematics, Nova Southeastern University, Fort Lauderdale, Florida, USA
| | - Clemencia Pinilla
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota, USA
| | - Michael S. Kriss
- Division of Gastroenterology and Hepatology, Department of Medicine, Aurora, Colorado, USA
| | - David J. Orlicky
- Department of Pathology, University of Colorado Anschutz Medical Campus. Aurora, Colorado, USA
| | - Beth A. Jirón Tamburini
- Division of Gastroenterology and Hepatology, Department of Medicine, Aurora, Colorado, USA
- Molecular Biology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Austin E. Gillen
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus. Aurora, Colorado, USA
| | - Matthew A. Burchill
- Division of Gastroenterology and Hepatology, Department of Medicine, Aurora, Colorado, USA
- Molecular Biology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
8
|
Curren B, Ahmed T, Rashid RB, Sebina I, Al Amin Sikder M, Howard DR, Alorro M, Ullah MA, Bissell A, Rahman MM, Pearen MA, Ramm GA, Varelias A, Rose-John S, MacDonald KPA, Hoelzle R, Ó Cuív P, Spann KM, Dennis PG, Phipps S. A maternal high-fat diet predisposes to infant lung disease via increased neutrophil-mediated IL-6 trans-signaling. Cell Rep 2024; 43:114974. [PMID: 39535919 DOI: 10.1016/j.celrep.2024.114974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 09/24/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
A poor maternal diet during pregnancy predisposes the infant to severe lower respiratory tract infections (sLRIs), which, in turn, increases childhood asthma risk; however, the underlying mechanisms remain poorly understood. Here, we show that the offspring of high-fat diet (HFD)-fed mothers (HFD-reared pups) developed an sLRI following pneumovirus inoculation in early life and subsequent asthma in later life upon allergen exposure. Prior to infection, HFD-reared pups developed microbial dysbiosis and low-grade systemic inflammation (LGSI), characterized by hyperneutropoiesis in the liver and elevated inflammatory cytokine expression, most notably granulocyte-colony stimulating factor (G-CSF), interleukin-17A (IL-17A), IL-6 and soluble IL-6 receptor (sIL-6R) (indicative of IL-6 trans-signaling) in the circulation and multiple organs but most prominently the liver. Inhibition of IL-6 trans-signaling using sgp130Fc transgenic mice or via specific genetic deletion of IL-6Ra on neutrophils conferred protection against both diseases. Taken together, our findings suggest that a maternal HFD induces neonatal LGSI that predisposes to sLRI and subsequent asthma via neutrophil-mediated IL-6 trans-signaling.
Collapse
Affiliation(s)
- Bodie Curren
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, QLD 4072, Australia
| | - Tufael Ahmed
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, QLD 4000, Australia
| | - Ridwan B Rashid
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Ismail Sebina
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Md Al Amin Sikder
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, QLD 4072, Australia
| | - Daniel R Howard
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, QLD 4072, Australia
| | - Mariah Alorro
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Md Ashik Ullah
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Alec Bissell
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Muhammed Mahfuzur Rahman
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, QLD 4072, Australia
| | - Michael A Pearen
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Grant A Ramm
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, QLD 4072, Australia
| | - Antiopi Varelias
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, QLD 4000, Australia; Australian Infectious Diseases Research Centre, The University of Queensland, QLD 4072, Australia
| | - Stefan Rose-John
- Christian-Albrechts-Universität zu Kiel, Medical Faculty, Olshausenstraße 40, 24098 Kiel, Germany
| | - Kelli P A MacDonald
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, QLD 4072, Australia
| | - Robert Hoelzle
- School of Environment, The University of Queensland, QLD 4072, Australia
| | - Páraic Ó Cuív
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, QLD 4000, Australia
| | - Kirsten M Spann
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, QLD 4000, Australia
| | - Paul G Dennis
- Australian Infectious Diseases Research Centre, The University of Queensland, QLD 4072, Australia; School of Environment, The University of Queensland, QLD 4072, Australia
| | - Simon Phipps
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, QLD 4072, Australia; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, QLD 4000, Australia; Australian Infectious Diseases Research Centre, The University of Queensland, QLD 4072, Australia.
| |
Collapse
|
9
|
Mori T, Yoshio S, Kakazu E, Kanto T. Active role of the immune system in metabolic dysfunction-associated steatotic liver disease. Gastroenterol Rep (Oxf) 2024; 12:goae089. [PMID: 39411101 PMCID: PMC11479709 DOI: 10.1093/gastro/goae089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/19/2024] [Accepted: 09/10/2024] [Indexed: 10/19/2024] Open
Abstract
Non-alcoholic fatty liver disease, recently renamed metabolic dysfunction-associated steatotic liver disease (MASLD), is a complex multifactorial disease that progresses from steatohepatitis (MASH) to liver cirrhosis and liver cancer. Recent research has revealed that crosstalk between innate immune cells and hepatic parenchymal and non-parenchymal cells is involved in the pathogenesis of liver disease in MASLD/MASH. Of particular importance, novel inflammatory mechanisms, including macrophage diversity, neutrophil NETosis, B-cell biology, auto-reactive T cells, unconventional T cells, and dendritic cell-T cell interactions, are considered key drivers for disease progression. These mechanisms and factors are potential targets for the therapeutic intervention of MASLD/MASH. In this review, we focus on recent discoveries related to liver inflammation and discuss the role of innate immune cell subsets in MASLD/MASH.
Collapse
Affiliation(s)
- Taizo Mori
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Chiba, Japan
| | - Sachiyo Yoshio
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Chiba, Japan
| | - Eiji Kakazu
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Chiba, Japan
| | - Tatsuya Kanto
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Chiba, Japan
| |
Collapse
|
10
|
Liu W, You D, Lin J, Zou H, Zhang L, Luo S, Yuan Y, Wang Z, Qi J, Wang W, Ye X, Yang X, Deng Y, Teng F, Zheng X, Lin Y, Huang Z, Huang Y, Yang Z, Zhou X, Zhang Y, Chen R, Xu L, Li J, Yang W, Zhang H. SGLT2 inhibitor promotes ketogenesis to improve MASH by suppressing CD8 + T cell activation. Cell Metab 2024; 36:2245-2261.e6. [PMID: 39243758 DOI: 10.1016/j.cmet.2024.08.005] [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: 01/23/2023] [Revised: 02/26/2024] [Accepted: 08/09/2024] [Indexed: 09/09/2024]
Abstract
During the progression of metabolic dysfunction-associated steatohepatitis (MASH), the accumulation of auto-aggressive CD8+ T cells significantly contributes to liver injury and inflammation. Empagliflozin (EMPA), a highly selective inhibitor of sodium-glucose co-transporter 2 (SGLT2), exhibits potential therapeutic benefits for liver steatosis; however, the underlying mechanism remains incompletely elucidated. Here, we found that EMPA significantly reduced the hepatic accumulation of auto-aggressive CD8+ T cells and lowered granzyme B levels in mice with MASH. Mechanistically, EMPA increased β-hydroxybutyric acid by promoting the ketogenesis of CD8+ T cells via elevating 3-hydroxybutyrate dehydrogenase 1 (Bdh1) expression. The β-hydroxybutyric acid subsequently inhibited interferon regulatory factor 4 (Irf4), which is crucial for CD8+ T cell activation. Furthermore, the ablation of Bdh1 in T cells aggravated the manifestation of MASH and hindered the therapeutic efficacy of EMPA. Moreover, a case-control study also showed that SGLT2 inhibitor treatment repressed CD8+ T cell infiltration and improved liver injury in patients with MASH. In summary, our study indicates that SGLT2 inhibitors can target CD8+ T cells and may be an effective strategy for treating MASH.
Collapse
Affiliation(s)
- Wenhui Liu
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Endocrinology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Danming You
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiayang Lin
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Huren Zou
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lei Zhang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shenjian Luo
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Youwen Yuan
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiyi Wang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jingwen Qi
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Weiwei Wang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xueru Ye
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoyu Yang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yajuan Deng
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fei Teng
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaojun Zheng
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yuhao Lin
- Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhiwei Huang
- Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yan Huang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhi Yang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xuan Zhou
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yanan Zhang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ruxin Chen
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lingling Xu
- Department of Endocrinology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Jin Li
- Division of Endocrinology, Department of Medicine, The Second Hospital of Shanxi Medical University, Taiyuan, China.
| | - Wei Yang
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Huijie Zhang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, China; State Key Laboratory of Organ Failure Research, Guangzhou, China; Guangdong Provincial Key Laboratory of Cell Metabolic Homeostasis and Major Chronic Diseases, Guangzhou, China.
| |
Collapse
|
11
|
Matchett KP, Paris J, Teichmann SA, Henderson NC. Spatial genomics: mapping human steatotic liver disease. Nat Rev Gastroenterol Hepatol 2024; 21:646-660. [PMID: 38654090 DOI: 10.1038/s41575-024-00915-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/28/2024] [Indexed: 04/25/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD, formerly known as non-alcoholic fatty liver disease) is a leading cause of chronic liver disease worldwide. MASLD can progress to metabolic dysfunction-associated steatohepatitis (MASH, formerly known as non-alcoholic steatohepatitis) with subsequent liver cirrhosis and hepatocellular carcinoma formation. The advent of current technologies such as single-cell and single-nuclei RNA sequencing have transformed our understanding of the liver in homeostasis and disease. The next frontier is contextualizing this single-cell information in its native spatial orientation. This understanding will markedly accelerate discovery science in hepatology, resulting in a further step-change in our knowledge of liver biology and pathobiology. In this Review, we discuss up-to-date knowledge of MASLD development and progression and how the burgeoning field of spatial genomics is driving exciting new developments in our understanding of human liver disease pathogenesis and therapeutic target identification.
Collapse
Affiliation(s)
- Kylie P Matchett
- Centre for Inflammation Research, Institute for Regeneration and Repair, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, UK
| | - Jasmin Paris
- Centre for Inflammation Research, Institute for Regeneration and Repair, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, UK
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Cambridge, UK
- Department of Physics, Cavendish Laboratory, University of Cambridge, Cambridge, UK
| | - Neil C Henderson
- Centre for Inflammation Research, Institute for Regeneration and Repair, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, UK.
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
12
|
Abdelnabi MN, Hassan GS, Shoukry NH. Role of the type 3 cytokines IL-17 and IL-22 in modulating metabolic dysfunction-associated steatotic liver disease. Front Immunol 2024; 15:1437046. [PMID: 39156888 PMCID: PMC11327067 DOI: 10.3389/fimmu.2024.1437046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/12/2024] [Indexed: 08/20/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) comprises a spectrum of liver diseases that span simple steatosis, metabolic dysfunction-associated steatohepatitis (MASH) and fibrosis and may progress to cirrhosis and cancer. The pathogenesis of MASLD is multifactorial and is driven by environmental, genetic, metabolic and immune factors. This review will focus on the role of the type 3 cytokines IL-17 and IL-22 in MASLD pathogenesis and progression. IL-17 and IL-22 are produced by similar adaptive and innate immune cells such as Th17 and innate lymphoid cells, respectively. IL-17-related signaling is upregulated during MASLD resulting in increased chemokines and proinflammatory cytokines in the liver microenvironment, enhanced recruitment of myeloid cells and T cells leading to exacerbation of inflammation and liver disease progression. IL-17 may also act directly by activating hepatic stellate cells resulting in increased fibrosis. In contrast, IL-22 is a pleiotropic cytokine with a dominantly protective signature in MASLD and is currently being tested as a therapeutic strategy. IL-22 also exhibits beneficial metabolic effects and abrogates MASH-related inflammation and fibrosis development via inducing the production of anti-oxidants and anti-apoptotic factors. A sex-dependent effect has been attributed to both cytokines, most importantly to IL-22 in MASLD or related conditions. Altogether, IL-17 and IL-22 are key effectors in MASLD pathogenesis and progression. We will review the role of these two cytokines and cells that produce them in the development of MASLD, their interaction with host factors driving MASLD including sexual dimorphism, and their potential therapeutic benefits.
Collapse
Affiliation(s)
- Mohamed N. Abdelnabi
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Ghada S. Hassan
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Naglaa H. Shoukry
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de médecine, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
13
|
Yong J, Wang D, Yu H. Machine learning-based integration of CD8 T cell-related gene signatures for comprehensive prognostic assessment in lung adenocarcinoma. Transl Cancer Res 2024; 13:3217-3241. [PMID: 39145093 PMCID: PMC11319961 DOI: 10.21037/tcr-23-2332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 06/02/2024] [Indexed: 08/16/2024]
Abstract
Background Lung adenocarcinoma (LUAD) stands as the most prevalent histological subtype of lung cancer, exhibiting heterogeneity in outcomes and diverse responses to therapy. CD8 T cells are consistently present throughout all stages of tumor development and play a pivotal role within the tumor microenvironment (TME). Our objective was to investigate the expression profiles of CD8 T cell marker genes, establish a prognostic risk model based on these genes in LUAD, and explore its relationship with immunotherapy response. Methods By leveraging the expression data and clinical records from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) cohorts, we identified 23 consensus prognostic genes. Employing ten machine-learning algorithms, we generated 101 combinations, ultimately selecting the optimal algorithm to construct an artificial intelligence-derived prognostic signature named riskScore. This selection was based on the average concordance index (C-index) across three testing cohorts. Results RiskScore emerged as an independent risk factor for overall survival (OS), progression-free interval (PFI), disease-free interval (DFI), and disease-specific survival (DSS) in LUAD. Notably, riskScore exhibited notably superior predictive accuracy compared to traditional clinical variables. Furthermore, we observed a positive correlation between the high-risk riskScore group and tumor-promoting biological functions, lower tumor mutational burden (TMB), lower neoantigen (NEO) load, and lower microsatellite instability (MSI) scores, as well as reduced immune cell infiltration and an increased probability of immune evasion within the TME. Of significance, the immunophenoscore (IPS) score displayed significant differences among risk subgroups, and riskScore effectively stratified patients in the IMvigor210 and GSE135222 immunotherapy cohort based on their survival outcomes. Additionally, we identified potential drugs that could target specific risk subgroups. Conclusions In summary, riskScore demonstrates its potential as a robust and promising tool for guiding clinical management and tailoring individualized treatments for LUAD patients.
Collapse
Affiliation(s)
- Jing Yong
- Department of Pharmacy, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Dongdong Wang
- Department of Oncology, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, The First People’s Hospital of Yancheng, Yancheng, China
| | - Huiming Yu
- Outpatient Dispensary for Chinese Traditional Medicine, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, The First People’s Hospital of Yancheng, Yancheng, China
| |
Collapse
|
14
|
Lambooij JM, Tak T, Zaldumbide A, Guigas B. OMIP-104: A 30-color spectral flow cytometry panel for comprehensive analysis of immune cell composition and macrophage subsets in mouse metabolic organs. Cytometry A 2024; 105:493-500. [PMID: 38651815 DOI: 10.1002/cyto.a.24845] [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: 12/23/2023] [Revised: 04/05/2024] [Accepted: 04/10/2024] [Indexed: 04/25/2024]
Abstract
Obesity-induced chronic low-grade inflammation, also known as metaflammation, results from alterations of the immune response in metabolic organs and contributes to the development of fatty liver diseases and type 2 diabetes. The diversity of tissue-resident leukocytes involved in these metabolic dysfunctions warrants an in-depth immunophenotyping in order to elucidate disease etiology. Here, we present a 30-color, full spectrum flow cytometry panel, designed to (i) identify the major innate and adaptive immune cell subsets in murine liver and white adipose tissues and (ii) discriminate various tissue-specific myeloid subsets known to contribute to the development of metabolic dysfunctions. This panel notably allows for distinguishing embryonically-derived liver-resident Kupffer cells from newly recruited monocyte-derived macrophages and KCs. Furthermore, several adipose tissue macrophage (ATM) subsets, including perivascular macrophages, lipid-associated macrophages, and pro-inflammatory CD11c+ ATMs, can also be identified. Finally, the panel includes cell-surface markers that have been associated with metabolic activation of different macrophage and dendritic cell subsets. Altogether, our spectral flow cytometry panel allows for an extensive immunophenotyping of murine metabolic tissues, with a particular focus on metabolically-relevant myeloid cell subsets, and can easily be adjusted to include various new markers if needed.
Collapse
Affiliation(s)
- Joost M Lambooij
- Leiden University Center of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
- Department of Cell & Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Tamar Tak
- Flow Cytometry Core Facility, Leiden University Medical Center, Leiden, The Netherlands
| | - Arnaud Zaldumbide
- Department of Cell & Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Bruno Guigas
- Leiden University Center of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
15
|
Tzeng HT, Lee WC. Impact of Transgenerational Nutrition on Nonalcoholic Fatty Liver Disease Development: Interplay between Gut Microbiota, Epigenetics and Immunity. Nutrients 2024; 16:1388. [PMID: 38732634 PMCID: PMC11085251 DOI: 10.3390/nu16091388] [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: 03/15/2024] [Revised: 04/25/2024] [Accepted: 05/01/2024] [Indexed: 05/13/2024] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) has emerged as the most prevalent pediatric liver disorder, primarily attributed to dietary shifts in recent years. NAFLD is characterized by the accumulation of lipid species in hepatocytes, leading to liver inflammation that can progress to steatohepatitis, fibrosis, and cirrhosis. Risk factors contributing to NAFLD encompass genetic variations and metabolic disorders such as obesity, diabetes, and insulin resistance. Moreover, transgenerational influences, resulting in an imbalance of gut microbial composition, epigenetic modifications, and dysregulated hepatic immune responses in offspring, play a pivotal role in pediatric NAFLD development. Maternal nutrition shapes the profile of microbiota-derived metabolites in offspring, exerting significant influence on immune system regulation and the development of metabolic syndrome in offspring. In this review, we summarize recent evidence elucidating the intricate interplay between gut microbiota, epigenetics, and immunity in fetuses exposed to maternal nutrition, and its impact on the onset of NAFLD in offspring. Furthermore, potential therapeutic strategies targeting this network are also discussed.
Collapse
Affiliation(s)
- Hong-Tai Tzeng
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan;
| | - Wei-Chia Lee
- Division of Urology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 33332, Taiwan
| |
Collapse
|
16
|
Zhang L, Shi Y, Liang B, Li X. An overview of the cholesterol metabolism and its proinflammatory role in the development of MASLD. Hepatol Commun 2024; 8:e0434. [PMID: 38696365 PMCID: PMC11068152 DOI: 10.1097/hc9.0000000000000434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/05/2024] [Indexed: 05/04/2024] Open
Abstract
Cholesterol is an essential lipid molecule in mammalian cells. It is not only involved in the formation of cell membranes but also serves as a raw material for the synthesis of bile acids, vitamin D, and steroid hormones. Additionally, it acts as a covalent modifier of proteins and plays a crucial role in numerous life processes. Generally, the metabolic processes of cholesterol absorption, synthesis, conversion, and efflux are strictly regulated. Excessive accumulation of cholesterol in the body is a risk factor for metabolic diseases such as cardiovascular disease, type 2 diabetes, and metabolic dysfunction-associated steatotic liver disease (MASLD). In this review, we first provide an overview of the discovery of cholesterol and the fundamental process of cholesterol metabolism. We then summarize the relationship between dietary cholesterol intake and the risk of developing MASLD, and also the animal models of MASLD specifically established with a cholesterol-containing diet. In the end, the role of cholesterol-induced inflammation in the initiation and development of MASLD is discussed.
Collapse
Affiliation(s)
- Linqiang Zhang
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Yongqiong Shi
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Bin Liang
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
| | - Xi Li
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
17
|
Li Z, Wang S, Xu Q, Su X, Wang Y, Wang L, Zhang Y. The double roles of T cell-mediated immune response in the progression of MASLD. Biomed Pharmacother 2024; 173:116333. [PMID: 38479177 DOI: 10.1016/j.biopha.2024.116333] [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: 01/07/2024] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/27/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease(MASLD), formerly known as non-alcoholic fatty liver disease(NAFLD), has become a major cause of chronic liver disease and a significant risk factor for hepatocellular carcinoma, which poses a huge burden on global public health and economy. MASLD includes steatotic liver disease, steatohepatitis, and cirrhosis, and the latter two cause great harm to human health and life, even complicated with liver cancer. Immunologic mechanism plays a major role in promoting its development into hepatitis and cirrhosis. Now more and more evidences show that T cells play an important role in the progression of MASLD. In this review, we discuss the double roles of T cells in MASLD from the perspective of T cell response pathways, as well as new evidences regarding the possible application of immunomodulatory therapy in MASH.
Collapse
Affiliation(s)
- Zigan Li
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| | - Shujun Wang
- Department of Medical Parasitology, Wannan Medical College, Wuhu 241000, China
| | - Qinchen Xu
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| | - Xin Su
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| | - Yunshan Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province 250021, China
| | - Lina Wang
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China.
| | - Yong Zhang
- Shandong Provincial Third Hospital Affiliated to Shandong University, Jinan, Shandong Province 250031, China.
| |
Collapse
|
18
|
Topchieva LV, Kurbatova IV, Dudanova OP, Vasileva AV, Zhulai GA. Immune cell balance as potential biomarker of progressing non-alcoholic fatty liver disease. GENES & CELLS 2024; 19:105-125. [DOI: 10.17816/gc610252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a widespread chronic, slowly progressive metabolic multifactorial disease. It is represented by several clinical and morphological forms: steatosis, nonalcoholic steatohepatitis (NASH) (with or without fibrosis), and liver cirrhosis. The search for minimally invasive and cost-effective biomarkers of NAFLD is a key task in the diagnosis, staging of progression, and long-term monitoring of NAFLD. This article discusses the possibility of using immune cell balance as potential minimally invasive peripheral markers of NAFLD progression. In the progression of NASH from steatosis to fibrosis and cirrhosis, inflammation plays an important role because of the activation of Kupffer cells and increased migration of monocytes, dendritic cells, neutrophils, and activated T lymphocytes into the tissues. Macrophages originating from monocytes, with NASH progression, gradually begin to prevail over the pool of resident macrophages. The risk of NASH and fibrosis development in patients with NAFLD increases with the ratio of neutrophils/lymphocytes in the liver. An increase in the Th17 cell count and a decrease in T-regulatory cell count can contribute to increased hepatic steatosis and inflammation development in NAFLD and accelerate the transition from simple steatosis to steatohepatitis and fibrosis. Information on the participation of noncoding RNAs in the regulation of the balance of immune cells in NAFLD is presented, which also allows us to consider them as additional, along with cellular, markers of disease progression.
Collapse
|
19
|
Niedecker RW, Delaney JA, Doyle MF, Sparks AD, Sitlani CM, Buzkova P, Zeb I, Tracy RP, Psaty BM, Budoff MJ, Olson NC. Investigating peripheral blood monocyte and T-cell subsets as non-invasive biomarkers for asymptomatic hepatic steatosis: results from the Multi-Ethnic Study of Atherosclerosis. Front Immunol 2024; 15:1243526. [PMID: 38596669 PMCID: PMC11002077 DOI: 10.3389/fimmu.2024.1243526] [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: 06/20/2023] [Accepted: 03/11/2024] [Indexed: 04/11/2024] Open
Abstract
Background Circulating immune cells have gained interest as biomarkers of hepatic steatosis. Data on the relationships between immune cell subsets and early-stage steatosis in population-based cohorts are limited. Methods This study included 1,944 asymptomatic participants of the Multi-Ethnic Study of Atherosclerosis (MESA) with immune cell phenotyping and computed tomography measures of liver fat. Participants with heavy alcohol use were excluded. A liver-to-spleen ratio Hounsfield units (HU) <1.0 and liver attenuation <40 HU were used to diagnose liver fat presence and >30% liver fat content, respectively. Logistic regression estimated cross-sectional associations of immune cell subsets with liver fat parameters adjusted for risk factors. We hypothesized that higher proportions of non-classical monocytes, Th1, Th17, and memory CD4+ T cells, and lower proportions of classical monocytes and naive CD4+ T cells, were associated with liver fat. Exploratory analyses evaluated additional immune cell phenotypes (n = 19). Results None of the hypothesized cells were associated with presence of liver fat. Higher memory CD4+ T cells were associated with >30% liver fat content, but this was not significant after correction for multiple hypothesis testing (odds ratio (OR): 1.31, 95% confidence interval (CI): 1.03, 1.66). In exploratory analyses unadjusted for multiple testing, higher proportions of CD8+CD57+ T cells were associated with liver fat presence (OR: 1.21, 95% CI: 1.02, 1.44) and >30% liver fat content (OR: 1.34, 95% CI: 1.07, 1.69). Conclusions Higher circulating memory CD4+ T cells may reflect liver fat severity. CD8+CD57+ cells were associated with liver fat presence and severity, but replication of findings is required.
Collapse
Affiliation(s)
- Rhys W. Niedecker
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, United States
| | - Joseph A. Delaney
- General Internal Medicine, University of Washington, Seattle, WA, United States
| | - Margaret F. Doyle
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, United States
| | - Andrew D. Sparks
- Department of Medical Biostatistics, Larner College of Medicine, University of Vermont, Burlington, VT, United States
| | - Colleen M. Sitlani
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Petra Buzkova
- Department of Biostatistics, University of Washington School of Public Health, Seattle, WA, United States
| | - Irfan Zeb
- Department of Medicine, West Virginia University Heart and Vascular Institute, Morgantown, WV, United States
| | - Russell P. Tracy
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, United States
- Department of Biochemistry, Larner College of Medicine, University of Vermont, Burlington, VT, United States
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, United States
- Department of Epidemiology, University of Washington, Seattle, WA, United States
- Department of Health Systems and Population Health, University of Washington, Seattle, WA, United States
| | - Matthew J. Budoff
- Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Nels C. Olson
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, United States
| |
Collapse
|
20
|
Pipitone RM, Lupo G, Zito R, Javed A, Petta S, Pennisi G, Grimaudo S. The PD-1/PD-L1 Axis in the Biology of MASLD. Int J Mol Sci 2024; 25:3671. [PMID: 38612483 PMCID: PMC11011676 DOI: 10.3390/ijms25073671] [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: 01/15/2024] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 04/14/2024] Open
Abstract
Metabolic Dysfunction-Associated Steatotic Liver (MASL), previously named nonalcoholic fatty liver (NAFL), is a multifactorial disease in which metabolic, genetic, and environmental risk factors play a predominant role. Obesity and type 2 diabetes act as triggers of the inflammatory response, which contributes to the progression of MASL to Metabolic Dysfunction-Associated Steatohepatitis and the development of hepatocellular carcinoma. In the liver, several parenchymal, nonparenchymal, and immune cells maintain immunological homeostasis, and different regulatory pathways balance the activation of the innate and adaptative immune system. PD-1/PD-L1 signaling acts, in the maintenance of the balance between the immune responses and the tissue immune homeostasis, promoting self-tolerance through the modulation of activated T cells. Recently, PD-1 has received much attention for its roles in inducing an exhausted T cells phenotype, promoting the tumor escape from immune responses. Indeed, in MASLD, the excessive fat accumulation dysregulates the immune system, increasing cytotoxic lymphocytes and decreasing their cytolytic activity. In this context, T cells exacerbate liver damage and promote tumor progression. The aim of this review is to illustrate the main pathogenetic mechanisms by which the immune system promotes the progression of MASLD and the transition to HCC, as well as to discuss the possible therapeutic applications of PD-1/PD-L1 target therapy to activate T cells and reinvigorate immune surveillance against cancer.
Collapse
|
21
|
Burtis AEC, DeNicola DMC, Ferguson ME, Santos RG, Pinilla C, Kriss MS, Orlicky DJ, Tamburini BAJ, Gillen AE, Burchill MA. Antigen-driven CD8 + T cell clonal expansion is a prominent feature of MASH in humans and mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.20.583964. [PMID: 38562766 PMCID: PMC10983976 DOI: 10.1101/2024.03.20.583964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Background and Aims Chronic liver disease due to metabolic dysfunction-associated steatohepatitis (MASH) is a rapidly increasing global epidemic. MASH progression is a consequence of the complex interplay between inflammatory insults and dysregulated hepatic immune responses. T lymphocytes have been shown to accumulate in the liver during MASH, but the cause and consequence of T cell accumulation in the liver remain unclear. Our study aimed to define the phenotype and T cell receptor diversity of T cells from human cirrhotic livers and an animal model of MASH to begin resolving their function in disease. Approach and Results In these studies, we evaluated differences in T cell phenotype in the context of liver disease we isolated liver resident T cell populations from individuals with cirrhosis and a murine model of MASH. Using both 5' single cell sequencing and flow cytometry we defined the phenotype and T cell receptor repertoire of liver resident T cells during health and disease. Conclusions MASH-induced cirrhosis and diet-induced MASH in mice resulted in the accumulation of activated and clonally expanded T cells in the liver. The clonally expanded T cells in the liver expressed markers of chronic antigenic stimulation, including PD1 , TIGIT and TOX . Overall, this study establishes for the first time that T cells undergo antigen-dependent clonal expansion and functional differentiation during the progression of MASH. These studies could lead to the identification of potential antigenic targets that drive T cell activation, clonal expansion, and recruitment to the liver during MASH.
Collapse
|
22
|
Lei Z, Yu J, Wu Y, Shen J, Lin S, Xue W, Mao C, Tang R, Sun H, Qi X, Wang X, Xu L, Wei C, Wang X, Chen H, Hao P, Yin W, Zhu J, Li Y, Wu Y, Liu S, Liang H, Chen X, Su C, Zhou S. CD1d protects against hepatocyte apoptosis in non-alcoholic steatohepatitis. J Hepatol 2024; 80:194-208. [PMID: 38438948 DOI: 10.1016/j.jhep.2023.10.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 10/02/2023] [Accepted: 10/17/2023] [Indexed: 03/06/2024]
Abstract
BACKGROUND & AIMS Hepatocyte apoptosis, a well-defined form of cell death in non-alcoholic steatohepatitis (NASH), is considered the primary cause of liver inflammation and fibrosis. However, the mechanisms underlying the regulation of hepatocyte apoptosis in NASH remain largely unclear. We explored the anti-apoptotic effect of hepatocyte CD1d in NASH. METHODS Hepatocyte CD1d expression was analyzed in patients with NASH and mouse models. Hepatocyte-specific gene overexpression or knockdown and anti-CD1d crosslinking were used to investigate the anti-apoptotic effect of hepatocyte CD1d on lipotoxicity-, Fas-, and concanavalin (ConA)-mediated liver injuries. A high-fat diet, a methionine-choline-deficient diet, a Fas agonist, and ConA were used to induce lipotoxic and/or apoptotic liver injuries. Palmitic acid was used to mimic lipotoxicity-induced apoptosis in vitro. RESULTS We identified a dramatic decrease in CD1d expression in hepatocytes of patients with NASH and mouse models. Hepatocyte-specific CD1d overexpression and knockdown experiments collectively demonstrated that hepatocyte CD1d protected against hepatocyte apoptosis and alleviated hepatic inflammation and injuries in NASH mice. Furthermore, decreased JAK2-STAT3 signaling was observed in NASH patient livers. Mechanistically, anti-CD1d crosslinking on hepatocytes induced tyrosine phosphorylation of the CD1d cytoplasmic tail, leading to the recruitment and phosphorylation of JAK2. Phosphorylated JAK2 activated STAT3 and subsequently reduced apoptosis in hepatocytes, which was associated with an increase in anti-apoptotic effectors (Bcl-xL and Mcl-1) and a decrease in pro-apoptotic effectors (cleaved-caspase 3/7). Moreover, anti-CD1d crosslinking effectively protected against Fas- or ConA-mediated hepatocyte apoptosis and liver injury in mice. CONCLUSIONS Our study uncovered a previously unrecognized anti-apoptotic CD1d-JAK2-STAT3 axis in hepatocytes that conferred hepatoprotection and highlighted the potential of hepatocyte CD1d-directed therapy for liver injury and fibrosis in NASH, as well as in other liver diseases associated with hepatocyte apoptosis. IMPACT AND IMPLICATIONS Excessive and/or sustained hepatocyte apoptosis is critical in driving liver inflammation and injury. The mechanisms underlying the regulation of hepatocyte apoptosis in non-alcoholic steatohepatitis (NASH) remain largely unclear. Here, we found that CD1d expression in hepatocytes substantially decreases and negatively correlates with the severity of liver injury in patients with NASH. We further revealed a previously unrecognized anti-apoptotic CD1d-JAK2-STAT3 signaling axis in hepatocytes, which confers significant protection against liver injury in NASH and acute liver diseases. Thus, hepatocyte CD1d-targeted therapy could be a promising strategy to manipulate liver injury in both NASH and other hepatocyte apoptosis-related liver diseases.
Collapse
Affiliation(s)
- Zhigang Lei
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiaojiao Yu
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yu Wu
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junyao Shen
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shibo Lin
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Weijie Xue
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chenxu Mao
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Rui Tang
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Haoran Sun
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xin Qi
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaohong Wang
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lei Xu
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chuan Wei
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaowei Wang
- Department of Blood Transfusion, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hongbing Chen
- Department of Clinical Laboratory, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ping Hao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Wen Yin
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Jifeng Zhu
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yalin Li
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yi Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Shouguo Liu
- Center for Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hui Liang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaojun Chen
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Chuan Su
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Sha Zhou
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
23
|
Xue M, Dong L, Zhang H, Li Y, Qiu K, Zhao Z, Gao M, Han L, Chan AKN, Li W, Leung K, Wang K, Pokharel SP, Qing Y, Liu W, Wang X, Ren L, Bi H, Yang L, Shen C, Chen Z, Melstrom L, Li H, Timchenko N, Deng X, Huang W, Rosen ST, Tian J, Xu L, Diao J, Chen CW, Chen J, Shen B, Chen H, Su R. METTL16 promotes liver cancer stem cell self-renewal via controlling ribosome biogenesis and mRNA translation. J Hematol Oncol 2024; 17:7. [PMID: 38302992 PMCID: PMC10835888 DOI: 10.1186/s13045-024-01526-9] [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: 04/19/2023] [Accepted: 01/20/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND While liver cancer stem cells (CSCs) play a crucial role in hepatocellular carcinoma (HCC) initiation, progression, recurrence, and treatment resistance, the mechanism underlying liver CSC self-renewal remains elusive. We aim to characterize the role of Methyltransferase 16 (METTL16), a recently identified RNA N6-methyladenosine (m6A) methyltransferase, in HCC development/maintenance, CSC stemness, as well as normal hepatogenesis. METHODS Liver-specific Mettl16 conditional KO (cKO) mice were generated to assess its role in HCC pathogenesis and normal hepatogenesis. Hydrodynamic tail-vein injection (HDTVi)-induced de novo hepatocarcinogenesis and xenograft models were utilized to determine the role of METTL16 in HCC initiation and progression. A limiting dilution assay was utilized to evaluate CSC frequency. Functionally essential targets were revealed via integrative analysis of multi-omics data, including RNA-seq, RNA immunoprecipitation (RIP)-seq, and ribosome profiling. RESULTS METTL16 is highly expressed in liver CSCs and its depletion dramatically decreased CSC frequency in vitro and in vivo. Mettl16 KO significantly attenuated HCC initiation and progression, yet only slightly influenced normal hepatogenesis. Mechanistic studies, including high-throughput sequencing, unveiled METTL16 as a key regulator of ribosomal RNA (rRNA) maturation and mRNA translation and identified eukaryotic translation initiation factor 3 subunit a (eIF3a) transcript as a bona-fide target of METTL16 in HCC. In addition, the functionally essential regions of METTL16 were revealed by CRISPR gene tiling scan, which will pave the way for the development of potential inhibitor(s). CONCLUSIONS Our findings highlight the crucial oncogenic role of METTL16 in promoting HCC pathogenesis and enhancing liver CSC self-renewal through augmenting mRNA translation efficiency.
Collapse
Affiliation(s)
- Meilin Xue
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, 91016, USA
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lei Dong
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, 91016, USA
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, UT Southwestern Medical Center, Dallas, TX, 7539, USA
| | - Honghai Zhang
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, 91016, USA
| | - Yangchan Li
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, 91016, USA
- Department of Radiation Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Kangqiang Qiu
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Zhicong Zhao
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, 91016, USA
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Min Gao
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, 91016, USA
| | - Li Han
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, 91016, USA
- School of Pharmacy, China Medical University, Shenyang, 110001, Liaoning, China
| | - Anthony K N Chan
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, 91016, USA
| | - Wei Li
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, 91016, USA
| | - Keith Leung
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, 91016, USA
| | - Kitty Wang
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, 91016, USA
| | - Sheela Pangeni Pokharel
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, 91016, USA
| | - Ying Qing
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, 91016, USA
| | - Wei Liu
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, 91016, USA
| | - Xueer Wang
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, 91016, USA
| | - Lili Ren
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, 91016, USA
| | - Hongjie Bi
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, 91016, USA
| | - Lu Yang
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, 91016, USA
| | - Chao Shen
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, 91016, USA
| | - Zhenhua Chen
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, 91016, USA
| | - Laleh Melstrom
- Division of Surgical Oncology, Department of Surgery, Beckman Research Institute of City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Hongzhi Li
- Department of Molecular Medicine, City of Hope National Medical Center, Duarte, CA, 91016, USA
| | - Nikolai Timchenko
- Division of General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Xiaolan Deng
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, 91016, USA
| | - Wendong Huang
- Department of Diabetes Complications and Metabolism, Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
- Graduate School of Biological Science, City of Hope, Duarte, CA, 91010, USA
| | - Steven T Rosen
- City of Hope Comprehensive Cancer Center, City of Hope, Duarte, CA, 91010, USA
| | - Jingyan Tian
- State Key Laboratory of Medical Genomics, Clinical Trial Center, Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lin Xu
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, UT Southwestern Medical Center, Dallas, TX, 7539, USA
| | - Jiajie Diao
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Chun-Wei Chen
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, 91016, USA
- City of Hope Comprehensive Cancer Center, City of Hope, Duarte, CA, 91010, USA
| | - Jianjun Chen
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, 91016, USA
- City of Hope Comprehensive Cancer Center, City of Hope, Duarte, CA, 91010, USA
- Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, 91010, USA
| | - Baiyong Shen
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Hao Chen
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Rui Su
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, 91016, USA.
- City of Hope Comprehensive Cancer Center, City of Hope, Duarte, CA, 91010, USA.
| |
Collapse
|
24
|
Hebbandi Nanjundappa R, Shao K, Krishnamurthy P, Gershwin ME, Leung PSC, Sokke Umeshappa C. Invariant natural killer T cells in autoimmune cholangiopathies: Mechanistic insights and therapeutic implications. Autoimmun Rev 2024; 23:103485. [PMID: 38040101 DOI: 10.1016/j.autrev.2023.103485] [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: 11/07/2023] [Accepted: 11/23/2023] [Indexed: 12/03/2023]
Abstract
Invariant natural killer T cells (iNKT cells) constitute a specialized subset of lymphocytes that bridges innate and adaptive immunity through a combination of traits characteristic of both conventional T cells and innate immune cells. iNKT cells are characterized by their invariant T cell receptors and discerning recognition of lipid antigens, which are presented by the non-classical MHC molecule, CD1d. Within the hepatic milieu, iNKT cells hold heightened prominence, contributing significantly to the orchestration of organ homeostasis. Their unique positioning to interact with diverse cellular entities, ranging from epithelial constituents like hepatocytes and cholangiocytes to immunocytes including Kupffer cells, B cells, T cells, and dendritic cells, imparts them with potent immunoregulatory abilities. Emergering knowledge of liver iNKT cells subsets enable to explore their therapeutic potential in autoimmne liver diseases. This comprehensive review navigates the landscape of iNKT cell investigations in immune-mediated cholangiopathies, with a particular focus on primary biliary cholangitis and primary sclerosing cholangitis, across murine models and human subjects to unravel the intricate involvements of iNKT cells in liver autoimmunity. Additionally, we also highlight the prospectives of iNKT cells as therapeutic targets in cholangiopathies. Modulation of the equilibrium between regulatory and proinflammatory iNKT subsets can be defining determinant in the dynamics of hepatic autoimmunity. This discernment not only enriches our foundational comprehension but also lays the groundwork for pioneering strategies to navigate the multifaceted landscape of liver autoimmunity.
Collapse
Affiliation(s)
| | - Kun Shao
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116023, China
| | - Prasanna Krishnamurthy
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - M Eric Gershwin
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, Davis, CA, United States.
| | - Patrick S C Leung
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, Davis, CA, United States
| | - Channakeshava Sokke Umeshappa
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada; Department of Pediatrics, IWK Research Center, Halifax, NS, Canada.
| |
Collapse
|
25
|
Parola M, Pinzani M. Liver fibrosis in NAFLD/NASH: from pathophysiology towards diagnostic and therapeutic strategies. Mol Aspects Med 2024; 95:101231. [PMID: 38056058 DOI: 10.1016/j.mam.2023.101231] [Citation(s) in RCA: 53] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/13/2023] [Accepted: 11/20/2023] [Indexed: 12/08/2023]
Abstract
Liver fibrosis, as an excess deposition of extracellular matrix (ECM) components, results from chronic liver injury as well as persistent activation of inflammatory response and of fibrogenesis. Liver fibrosis is a major determinant for chronic liver disease (CLD) progression and in the last two decades our understanding on the major molecular and cellular mechanisms underlying the fibrogenic progression of CLD has dramatically improved, boosting pre-clinical studies and clinical trials designed to find novel therapeutic approaches. From these studies several critical concepts have emerged, starting to reveal the complexity of the pro-fibrotic microenvironment which involves very complex, dynamic and interrelated interactions between different hepatic and extrahepatic cell populations. This review will offer first a recapitulation of established and novel pathophysiological basic principles and concepts by intentionally focus the attention on NAFLD/NASH, a metabolic-related form of CLD with a high impact on the general population and emerging as a leading cause of CLD worldwide. NAFLD/NASH-related pro-inflammatory and profibrogenic mechanisms will be analysed as well as novel information on cells, mediators and signalling pathways which have taken advantage from novel methodological approaches and techniques (single cell genomics, imaging mass cytometry, novel in vitro two- and three-dimensional models, etc.). We will next offer an overview on recent advancement in diagnostic and prognostic tools, including serum biomarkers and polygenic scores, to support the analysis of liver biopsies. Finally, this review will provide an analysis of current and emerging therapies for the treatment of NAFLD/NASH patients.
Collapse
Affiliation(s)
- Maurizio Parola
- Dept. Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Corso Raffaello 30, 10125, Torino, Italy.
| | - Massimo Pinzani
- UCL Institute for Liver and Digestive Health, Division of Medicine - Royal Free Hospital, London, NW32PF, United Kingdom.
| |
Collapse
|
26
|
Ali M, Benfante V, Di Raimondo D, Salvaggio G, Tuttolomondo A, Comelli A. Recent Developments in Nanoparticle Formulations for Resveratrol Encapsulation as an Anticancer Agent. Pharmaceuticals (Basel) 2024; 17:126. [PMID: 38256959 PMCID: PMC10818631 DOI: 10.3390/ph17010126] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/13/2024] [Accepted: 01/15/2024] [Indexed: 01/24/2024] Open
Abstract
Resveratrol is a polyphenolic compound that has gained considerable attention in the past decade due to its multifaceted therapeutic potential, including anti-inflammatory and anticancer properties. However, its anticancer efficacy is impeded by low water solubility, dose-limiting toxicity, low bioavailability, and rapid hepatic metabolism. To overcome these hurdles, various nanoparticles such as organic and inorganic nanoparticles, liposomes, polymeric nanoparticles, dendrimers, solid lipid nanoparticles, gold nanoparticles, zinc oxide nanoparticles, zeolitic imidazolate frameworks, carbon nanotubes, bioactive glass nanoparticles, and mesoporous nanoparticles were employed to deliver resveratrol, enhancing its water solubility, bioavailability, and efficacy against various types of cancer. Resveratrol-loaded nanoparticle or resveratrol-conjugated nanoparticle administration exhibits excellent anticancer potency compared to free resveratrol. This review highlights the latest developments in nanoparticle-based delivery systems for resveratrol, focusing on the potential to overcome limitations associated with the compound's bioavailability and therapeutic effectiveness.
Collapse
Affiliation(s)
- Muhammad Ali
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy;
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy; (D.D.R.); (A.T.)
| | - Viviana Benfante
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy;
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy; (D.D.R.); (A.T.)
| | - Domenico Di Raimondo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy; (D.D.R.); (A.T.)
| | - Giuseppe Salvaggio
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy;
| | - Antonino Tuttolomondo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy; (D.D.R.); (A.T.)
| | - Albert Comelli
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy;
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| |
Collapse
|
27
|
Adams VR, Collins LB, Williams TI, Holmes J, Hess P, Atkins HM, Scheidemantle G, Liu X, Lodge M, Johnson AJ, Kennedy A. Myeloid cell MHC I expression drives CD8 + T cell activation in nonalcoholic steatohepatitis. Front Immunol 2024; 14:1302006. [PMID: 38274832 PMCID: PMC10808415 DOI: 10.3389/fimmu.2023.1302006] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 11/28/2023] [Indexed: 01/27/2024] Open
Abstract
Background & aims Activated CD8+ T cells are elevated in Nonalcoholic steatohepatitis (NASH) and are important for driving fibrosis and inflammation. Despite this, mechanisms of CD8+ T cell activation in NASH are largely limited. Specific CD8+ T cell subsets may become activated through metabolic signals or cytokines. However, studies in NASH have not evaluated the impact of antigen presentation or the involvement of specific antigens. Therefore, we determined if activated CD8+ T cells are dependent on MHC class I expression in NASH to regulate fibrosis and inflammation. Methods We used H2Kb and H2Db deficient (MHC I KO), Kb transgenic mice, and myeloid cell Kb deficient mice (LysM Kb KO) to investigate how MHC class I impacts CD8+ T cell function and NASH. Flow cytometry, gene expression, and histology were used to examine hepatic inflammation and fibrosis. The hepatic class I immunopeptidome was evaluated by mass spectrometry. Results In NASH, MHC class I isoform H2Kb was upregulated in myeloid cells. MHC I KO demonstrated protective effects against NASH-induced inflammation and fibrosis. Kb mice exhibited increased fibrosis in the absence of H2Db while LysM Kb KO mice showed protection against fibrosis but not inflammation. H2Kb restricted peptides identified a unique NASH peptide Ncf2 capable of CD8+ T cell activation in vitro. The Ncf2 peptide was not detected during fibrosis resolution. Conclusion These results suggest that activated hepatic CD8+ T cells are dependent on myeloid cell MHC class I expression in diet induced NASH to promote inflammation and fibrosis. Additionally, our studies suggest a role of NADPH oxidase in the production of Ncf2 peptide generation.
Collapse
Affiliation(s)
- Victoria R. Adams
- Department of Molecular and Structural Biochemistry, NC State University, Raleigh, NC, United States
| | - Leonard B. Collins
- Molecular Education, Technology and Research Innovation Center (METRIC), NC State University, Raleigh, NC, United States
| | - Taufika Islam Williams
- Molecular Education, Technology and Research Innovation Center (METRIC), NC State University, Raleigh, NC, United States
- Department of Chemistry, NC State University, Raleigh, NC, United States
| | - Jennifer Holmes
- College of Veterinary Medicine, NC State University, Raleigh, NC, United States
| | - Paul Hess
- College of Veterinary Medicine, NC State University, Raleigh, NC, United States
| | - Hannah M. Atkins
- Center for Human Health and Environment, NC State University, Raleigh, NC, United States
- Division of Comparative Medicine, UNC Chapel Hill, Chapel Hill, NC, United States
| | - Grace Scheidemantle
- Department of Molecular and Structural Biochemistry, NC State University, Raleigh, NC, United States
| | - Xiaojing Liu
- Department of Molecular and Structural Biochemistry, NC State University, Raleigh, NC, United States
| | - Mareca Lodge
- Department of Molecular and Structural Biochemistry, NC State University, Raleigh, NC, United States
| | - Aaron J. Johnson
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | - Arion Kennedy
- Department of Molecular and Structural Biochemistry, NC State University, Raleigh, NC, United States
| |
Collapse
|
28
|
Bhattacharjee J, Beaton G, Ravula SB, Lee SJ, Bacon KB, Jenkinson CP, Warren M, Tucci FC, Kohli R. Lysophosphatidic acid receptor 1 antagonist (EPGN2154) causes regression of NASH in preclinical NASH models. Hepatol Commun 2023; 7:e0323. [PMID: 37994050 PMCID: PMC10666985 DOI: 10.1097/hc9.0000000000000323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/28/2023] [Indexed: 11/24/2023] Open
Abstract
BACKGROUND NASH causes a tremendous health care burden in the United States. A glucagon-like peptide-1 agonist, semaglutide (Sema), treatment resulted in hepatic steatosis reduction in clinical trials of NASH. Lysophosphatidic acid receptor 1 antagonists are known to have antifibrotic effects in several organs. We tested Sema and a novel lysophosphatidic acid receptor 1 antagonist, EPGN2154, individually and in combination to evaluate their efficacy for NASH remission in preclinical models. METHODS In the present study, we used (1) C57Bl6/J wild-type mice fed on a high-fat, high-carbohydrate (HFHC) diet for 16 weeks and (2) leptin-deficient mice (ob/ob) fed on an Amylin liver NASH diet for 16 weeks. After 16 weeks, the mice were randomly distributed in equal numbers in (1) no-drug, (2) EPGN2154, (3) Sema, and (4) EPGN2154+Sema treatment groups for 8 additional weeks at a dosage of 10 mg/kg body weight for EPGN2154 (oral gavage, 5 days a week) and 6.17 μg/kg body weight of Sema (subcutaneous injection every alternate day, 3 days a week). RESULTS In the wild-type-high-fat, high-carbohydrate model, we observed the most body weight loss in the EPGN2154+Sema combination group compared to the other treatment groups. All groups led to a significant reduction in alanine transaminase levels when compared to high-fat, high-carbohydrate-fed wild type. However, no significant difference in alanine transaminase levels was observed among the treatment groups. In the ob/ob mice study, Sema did not cause body weight loss. Moreover, the EPGN2154 and the combination groups had a lower NAFLD Activity Score and incidence of advanced-stage hepatic fibrosis than the Sema group. CONCLUSIONS EPGN2154 demonstrated a hepato-protective effect independent of body weight loss in preclinical NASH models.
Collapse
Affiliation(s)
- Jashdeep Bhattacharjee
- Department of Gastroenterology, Hepatology and Nutrition, Children’s Hospital Los Angeles, Los Angeles, California, USA
| | | | | | | | | | | | - Mikako Warren
- Department of Pathology and Laboratory Medicine, Children’s Hospital Los Angeles, Los Angeles, California, USA
| | | | - Rohit Kohli
- Department of Gastroenterology, Hepatology and Nutrition, Children’s Hospital Los Angeles, Los Angeles, California, USA
| |
Collapse
|
29
|
Zhao Q, Wu J, Ding Y, Pang Y, Jiang C. Gut microbiota, immunity, and bile acid metabolism: decoding metabolic disease interactions. LIFE METABOLISM 2023; 2:load032. [PMID: 39872860 PMCID: PMC11749371 DOI: 10.1093/lifemeta/load032] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 07/14/2023] [Accepted: 07/21/2023] [Indexed: 01/03/2025]
Abstract
In recent decades, the global prevalence of metabolic syndrome has surged, posing a significant public health challenge. Metabolic disorders, encompassing diabetes, obesity, nonalcoholic fatty liver disease, and polycystic ovarian syndrome, have been linked to alterations in the gut microbiota. Nonetheless, the connection between gut microbiota and host metabolic diseases warrants further investigation. In this review, we delve into the associations between various metabolic disorders and the gut microbiota, focusing on immune responses and bile acid (BA) metabolism. Notably, T helper cells, innate lymphoid cells, macrophages, and dendritic cells have been shown to modulate host metabolism through interactions with intestinal microorganisms and the release of cytokines. Furthermore, secondary BA metabolites, derived from the microbiota, are involved in the pathogenesis of metabolic diseases via the farnesoid X receptor and Takeda G protein-coupled receptor 5. By covering both aspects of this immune system-microorganism axis, we present a comprehensive overview of the roles played by the gut microbiota, microbiota-derived BA metabolites, and immune responses in metabolic diseases, as well as the interplay between these systems.
Collapse
Affiliation(s)
- Qixiang Zhao
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
- Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Jiayu Wu
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
- Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Yong Ding
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
- Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Yanli Pang
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
| | - Changtao Jiang
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
- Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
30
|
Sawada K, Chung H, Softic S, Moreno-Fernandez ME, Divanovic S. The bidirectional immune crosstalk in metabolic dysfunction-associated steatotic liver disease. Cell Metab 2023; 35:1852-1871. [PMID: 37939656 PMCID: PMC10680147 DOI: 10.1016/j.cmet.2023.10.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 10/13/2023] [Accepted: 10/13/2023] [Indexed: 11/10/2023]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is an unabated risk factor for end-stage liver diseases with no available therapies. Dysregulated immune responses are critical culprits of MASLD pathogenesis. Independent contributions from either the innate or adaptive arms of the immune system or their unidirectional interplay are commonly studied in MASLD. However, the bidirectional communication between innate and adaptive immune systems and its impact on MASLD remain insufficiently understood. Given that both innate and adaptive immune cells are indispensable for the development and progression of inflammation in MASLD, elucidating pathogenic contributions stemming from the bidirectional interplay between these two arms holds potential for development of novel therapeutics for MASLD. Here, we review the immune cell types and bidirectional pathways that influence the pathogenesis of MASLD and highlight potential pharmacologic approaches to combat MASLD based on current knowledge of this bidirectional crosstalk.
Collapse
Affiliation(s)
- Keisuke Sawada
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
| | - Hak Chung
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Samir Softic
- Department of Pediatrics and Gastroenterology, University of Kentucky, Lexington, KY 40536, USA; Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Maria E Moreno-Fernandez
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| | - Senad Divanovic
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|
31
|
Bai J, Zhu L, Mi W, Gao Z, Ouyang M, Sheng W, Song L, Bao L, Ma Y, Xu Y. Multiscale integrative analyses unveil immune-related diagnostic signature for the progression of MASLD. Hepatol Commun 2023; 7:e0298. [PMID: 37851406 PMCID: PMC10586828 DOI: 10.1097/hc9.0000000000000298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 08/25/2023] [Indexed: 10/19/2023] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a chronic liver disease prevalent worldwide, with an increasing incidence associated with obesity, diabetes, and metabolic syndrome. The progression of MASLD to metabolic dysfunction-associated steatohepatitis (MASH) poses a pressing health concern, highlighting the significance of accurately identifying MASLD and its progression to MASH as a primary challenge in the field. In this study, a systematic integration of 66 immune cell types was conducted. Comprehensive analyses were performed on bulk, single-cell RNA-Seq, and clinical data to investigate the immune cell types implicated in MASLD progression thoroughly. Multiple approaches, including immune infiltration, gene expression trend analysis, weighted gene coexpression network analysis, and 4 machine learning algorithms, were used to examine the dynamic changes in genes and immune cells during MASLD progression. C-X-C motif chemokine receptor 4 and dedicator of cytokinesis 8 have been identified as potential diagnostic biomarkers for MASLD progression. Furthermore, cell communication analysis at the single-cell level revealed that the involvement of C-X-C motif chemokine receptor 4 and dedicator of cytokinesis 8 in MASLD progression is mediated through their influence on T cells. Overall, our study identified vital immune cells and a 2-gene diagnostic signature for the progression of MASLD, providing a new perspective on the diagnosis and immune-related molecular mechanisms of MASLD. These findings have important implications for developing innovative diagnostic tools and therapies for MASLD.
Collapse
Affiliation(s)
- Jing Bai
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Lun Zhu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Wanqi Mi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Zhengzheng Gao
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| | - Minyue Ouyang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Wanlu Sheng
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| | - Lin Song
- College of Mongolian Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Lidao Bao
- Hohhot Mongolian Medicine of Traditional Chinese Medicine Hospital, Hohhot, China
| | - Yuheng Ma
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| | - Yingqi Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
- College of Mongolian Medicine, Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
32
|
Kumar V, Hertz M, Agro A, Byrne AJ. Type 1 invariant natural killer T cells in chronic inflammation and tissue fibrosis. Front Immunol 2023; 14:1260503. [PMID: 37818376 PMCID: PMC10561218 DOI: 10.3389/fimmu.2023.1260503] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/06/2023] [Indexed: 10/12/2023] Open
Abstract
Chronic tissue inflammation often results in fibrosis characterized by the accumulation of extracellular matrix components remodeling normal tissue architecture and function. Recent studies have suggested common immune mechanisms despite the complexity of the interactions between tissue-specific fibroblasts, macrophages, and distinct immune cell populations that mediate fibrosis in various tissues. Natural killer T (NKT) cells recognizing lipid antigens bound to CD1d molecules have been shown to play an important role in chronic inflammation and fibrosis. Here we review recent data in both experimental models and in humans that suggest a key role of type 1 invariant NKT (iNKT) cell activation in the progression of inflammatory cascades leading to recruitment of neutrophils and activation of the inflammasome, macrophages, fibroblasts, and, ultimately, fibrosis. Emerging evidence suggests that iNKT-associated mechanisms contribute to type 1, type 2 and type 3 immune pathways mediating tissue fibrosis, including idiopathic pulmonary fibrosis (IPF). Thus, targeting a pathway upstream of these immune mechanisms, such as the inhibition of iNKT activation, may be important in modulating various fibrotic conditions.
Collapse
Affiliation(s)
- Vipin Kumar
- Laboratory of Immune Regulation, Department of Medicine, University of California San Diego, La Jolla, CA, United States
- GRI Bio, La Jolla, CA, United States
| | | | | | - Adam J. Byrne
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- School of Medicine and Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| |
Collapse
|
33
|
Ishikawa H, Nagashima R, Kuno Y, Sasaki H, Kohda C, Iyoda M. Effects of NKT Cells on Metabolic Disorders Caused by High-Fat Diet Using CD1d-Knockout Mice. Diabetes Metab Syndr Obes 2023; 16:2855-2864. [PMID: 37744699 PMCID: PMC10517681 DOI: 10.2147/dmso.s428190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/14/2023] [Indexed: 09/26/2023] Open
Abstract
Purpose The purpose of this study was to investigate whether NKT cells play an important role in preventing or exacerbating diseases caused by high-fat diet (HFD) using CD1d-knockout (KO) mice which lack NKT cells. Methods Five-week-old male Balb/c (wild-type; WT) or CD1dKO mice were fed with control-diet (CTD) or HFD for 16 weeks. Results The present study revealed four main findings. First, CD1dKO mice were susceptible to obesity caused by HFD in comparison to WT mice. Second, clinical conditions of fatty liver caused by HFD were comparable between CD1dKO mice and WT mice. Third, HFD-fed WT mice showed high levels of serum biochemical markers, involved in lipid metabolisms, in comparison to WT mice fed a CTD. Notably, the serum concentrations of ALT, T-CHO, TG and HDL-C in CD1dKO mice fed a HFD were almost comparable to those of CD1dKO mice fed a CTD. Fourth, the expression of peroxisome proliferator-activated receptor (PPAR) γ, low-density lipoprotein receptor (LDLR), CD36 of epididymal adipose tissue enhanced and proprotein convertase subtilisin/kexin type (PCSK) 9 in serum decreased. Conclusion NKT cells were responsible for protection against HFD-induced obesity. However, CD1dKO mice were resistant to serum biochemical marker abnormalities after HFD feeding. One possible explanation is that the epididymal adipose tissue of CD1dKO mice could take up greater amounts of excess lipids in serum in comparison to WT mice.
Collapse
Affiliation(s)
- Hiroki Ishikawa
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, 142-8555, Japan
| | - Ryuichi Nagashima
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, 142-8555, Japan
| | - Yoshihiro Kuno
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, 142-8555, Japan
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, 142-8666, Japan
| | - Hiraku Sasaki
- Department of Health Science, Faculty of Health and Sports Science, Juntendo University, Inzai, Chiba, 270-1695, Japan
| | - Chikara Kohda
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, 142-8555, Japan
| | - Masayuki Iyoda
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, 142-8555, Japan
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, 142-8666, Japan
| |
Collapse
|
34
|
Zhong X, Lv M, Ma M, Huang Q, Hu R, Li J, Yi J, Sun J, Zhou X. State of CD8 + T cells in progression from nonalcoholic steatohepatitis to hepatocellular carcinoma: From pathogenesis to immunotherapy. Biomed Pharmacother 2023; 165:115131. [PMID: 37429231 DOI: 10.1016/j.biopha.2023.115131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/26/2023] [Accepted: 07/02/2023] [Indexed: 07/12/2023] Open
Abstract
With the obesity epidemic, nonalcoholic steatohepatitis (NASH) is emerging as the fastest growing potential cause of hepatocellular carcinoma (HCC). NASH has been demonstrated to establish a tumor-prone liver microenvironment where both innate and adaptive immune systems are involved. As the most typical anti-tumor effector, the cell function of CD8+ T cells is remodeled by chronic inflammation, metabolic alteration, lipid toxicity and oxidative stress in the liver microenvironment along the NASH to HCC transition. Unexpectedly, NASH may blunt the effect of immune checkpoint inhibitor therapy against HCC due to the dysregulated CD8+ T cells. Growing evidence has supported that NASH is likely to facilitate the state transition of CD8+ T cells with changes in cell motility, effector function, metabolic reprogramming and gene transcription according to single-cell sequencing. However, the mechanistic insight of CD8+ T cell states in the NASH-driven HCC is not comprehensive. Herein, we focus on the characterization of state phenotypes of CD8+ T cells with both functional and metabolic signatures in NASH-driven fibrosis and HCC. The NASH-specific CD8+ T cells are speculated to mainly have a dualist effect, where its aberrant activated phenotype sustains chronic inflammation in NASH but subsequently triggers its exhaustion in HCC. As the exploration of CD8+ T cells on the distribution and phenotypic shifts will provide a new direction for the intervention strategies against HCC, we also discuss the implications for targeting different phenotypes of CD8+ T cells, shedding light on the personalized immunotherapy for NASH-driven HCC.
Collapse
Affiliation(s)
- Xin Zhong
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China; Department of Liver Disease, the fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Minling Lv
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China; Department of Liver Disease, the fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - MengQing Ma
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China; Department of Liver Disease, the fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Qi Huang
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China; Department of Liver Disease, the fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Rui Hu
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China; Department of Liver Disease, the fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Jing Li
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China; Department of Liver Disease, the fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Jinyu Yi
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China; Department of Liver Disease, the fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Jialing Sun
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China; Department of Liver Disease, the fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Xiaozhou Zhou
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China; Department of Liver Disease, the fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China.
| |
Collapse
|
35
|
Abstract
The understanding of the mechanisms of liver fibrosis has been dominated by models in which chronic hepatocellular injury is the initiating step as is seen with viral infections. The increased prevalence of the metabolic syndrome, and the increases in liver fibrosis due to metabolic syndrome driven non-alcoholic steatohepatitis (NASH), has made it a priority to understand how this type of liver fibrosis is similar to, and different from, pure hepatocellular injury driven liver fibrosis. Both types of liver fibrosis have the transformation of the hepatic stellate cell (HSC) into a myofibroblast as a key step. In metabolic syndrome, there is little evidence that metabolite changes such as high levels of glucose and free fatty acids are directly inducing HSC transdifferentiation, however, metabolite changes may lead to reductions in immunomodulatory and hepatoprotective molecules such as lipoxins, resolvins and Interleukin (IL)-22. Cells of the innate immune system are known to be important intermediaries between hepatocellular damage and HSC transdifferentiation, primarily by producing cytokines such as transforming growth factor-β (TGF-β) and platelet derived growth factor (PDGF). Resident and infiltrating macrophages are the dominant innate immune cells, but others (dendritic cells, neutrophils, natural killer T cells and mucosal-associated invariant T cells) also have important roles in inducing and resolving liver fibrosis. CD8+ and CD4+ T cells of the adaptive immune system have been identified to have greater profibrotic roles than previously realised by inducing hepatocyte death (auto-aggressive CD8+T) cells and cytokines producing (TH17 producing CD4+T) cells. Finally, the cellular networks present in NASH fibrosis are being identified and suggest that once fibrosis has developed cell-to-cell communication is dominated by myofibroblasts autocrine signalling followed by communication with cholangiocytes and endothelial cells, with myofibroblast-hepatocyte, and myofibroblast-macrophage signalling having minor roles. Such information is essential to the development of antifibrotic strategies for different stages of fibrosis.
Collapse
Affiliation(s)
- Wajahat Mehal
- Section of Digestive Diseases, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
36
|
Li X, Lu Y, Liang X, Zhou X, Li D, Zhang Z, Niu Y, Liu S, Ye L, Zhang R. A new NASH model in aged mice with rapid progression of steatohepatitis and fibrosis. PLoS One 2023; 18:e0286257. [PMID: 37228085 DOI: 10.1371/journal.pone.0286257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/11/2023] [Indexed: 05/27/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has a high prevalence worldwide, with a significant proportion of patients progressing into non-alcoholic steatohepatitis (NASH) and further into cirrhosis and hepatocellular carcinoma (HCC). Most of the current animal models of NASH have limitations, such as incompatibility with human pathogenesis characteristics or long induction periods, which severely limit the development of new drugs and preclinical studies for NASH. We investigated the progression of NASH and fibrosis, as well as metabolic indicators, at different time points in aged mice induced by the Gubra Amylin NASH (GAN) diet, a high-fat, high-sugar, high-cholesterol diet, and attempted to establish a rapid and useful mouse model of NASH. Young and aged C57BL/6 mice were induced on a normal chow or GAN diet for 12 and 21 weeks, respectively. After 12 weeks of induction, aged mice developed NASH, including hepatic steatosis, lobular inflammation and hepatic ballooning, and the phenotype was more severe compared with young mice. After 21 weeks of induction, aged mice developed hepatic fibrosis, which greatly shortened the induction time compared with young mice. Furthermore, analysis of immune cell infiltration in the liver by flow cytometry elucidated the changes of multiple immune cells during the pathogenesis of NASH. These findings suggest that aged mice may develop NASH and fibrosis more rapidly under GAN diet induction, which may significantly shorten the period for preclinical studies of NASH.
Collapse
Affiliation(s)
- Xuecheng Li
- Biocytogen Pharmaceuticals (Beijing) Co., Ltd, Beijing, Daxing District, China
| | - Yi Lu
- Biocytogen Pharmaceuticals (Beijing) Co., Ltd, Beijing, Daxing District, China
| | - Xiaoshuang Liang
- Biocytogen Pharmaceuticals (Beijing) Co., Ltd, Beijing, Daxing District, China
| | - Xiaofei Zhou
- Biocytogen Pharmaceuticals (Beijing) Co., Ltd, Beijing, Daxing District, China
| | - Dirui Li
- Biocytogen Pharmaceuticals (Beijing) Co., Ltd, Beijing, Daxing District, China
| | - Zan Zhang
- Biocytogen Pharmaceuticals (Beijing) Co., Ltd, Beijing, Daxing District, China
| | - Yunchao Niu
- Biocytogen Pharmaceuticals (Beijing) Co., Ltd, Beijing, Daxing District, China
| | - Shuaishuai Liu
- Biocytogen Pharmaceuticals (Beijing) Co., Ltd, Beijing, Daxing District, China
| | - Ling Ye
- Biocytogen Pharmaceuticals (Beijing) Co., Ltd, Beijing, Daxing District, China
| | - Rufeng Zhang
- Biocytogen Pharmaceuticals (Beijing) Co., Ltd, Beijing, Daxing District, China
| |
Collapse
|
37
|
Zhu Z, Chen Y, Qin X, Liu S, Wang J, Ren H. Multidimensional landscape of non-alcoholic fatty liver disease-related disease spectrum uncovered by big omics data: Profiling evidence and new perspectives. SMART MEDICINE 2023; 2:e20220029. [PMID: 39188279 PMCID: PMC11236021 DOI: 10.1002/smmd.20220029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 02/22/2023] [Indexed: 08/28/2024]
Abstract
Characterized by hepatic lipid accumulation, non-alcoholic fatty liver disease (NAFLD) is a multifactorial metabolic disorder that could promote the progression of non-alcoholic steatohepatitis (NASH), cirrhosis, and hepatocellular carcinoma (HCC). Benefiting from recent advances in omics technologies, such as high-throughput sequencing, voluminous profiling data in HCC-integrated molecular science into clinical medicine helped clinicians with rational guidance for treatments. In this review, we conclude the majority of publicly available omics data on the NAFLD-related disease spectrum and bring up new insights to inspire next-generation therapeutics against this increasingly prevalent disease spectrum in the post-genomic era.
Collapse
Affiliation(s)
- Zhengyi Zhu
- Department of Hepatobiliary SurgeryAffiliated Drum Tower HospitalMedical SchoolNanjing UniversityNanjingChina
| | - Yuyan Chen
- Department of Hepatobiliary SurgeryAffiliated Drum Tower HospitalMedical SchoolNanjing UniversityNanjingChina
| | - Xueqian Qin
- Department of Hepatobiliary SurgeryAffiliated Drum Tower HospitalMedical SchoolNanjing UniversityNanjingChina
| | - Shujun Liu
- Department of Hepatobiliary SurgeryAffiliated Drum Tower HospitalMedical SchoolNanjing UniversityNanjingChina
| | - Jinglin Wang
- Department of Hepatobiliary SurgeryAffiliated Drum Tower HospitalMedical SchoolNanjing UniversityNanjingChina
| | - Haozhen Ren
- Department of Hepatobiliary SurgeryAffiliated Drum Tower HospitalMedical SchoolNanjing UniversityNanjingChina
| |
Collapse
|
38
|
Papanastasatou M, Verykokakis M. Innate-like T lymphocytes in chronic liver disease. Front Immunol 2023; 14:1114605. [PMID: 37006304 PMCID: PMC10050337 DOI: 10.3389/fimmu.2023.1114605] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/28/2023] [Indexed: 03/17/2023] Open
Abstract
In addition to its metabolic activities, it is now clear that the liver hosts a number of diverse immune cell types that control tissue homeostasis. Foremost among these are innate-like T lymphocytes, including natural killer T (NKT) and mucosal-associated innate T (MAIT) cells, which are a population of specialized T cells with innate characteristics that express semi-invariant T cell receptors with non-peptide antigen specificity. As primary liver residents, innate-like T cells have been associated with immune tolerance in the liver, but also with a number of hepatic diseases. Here, we focus on the biology of NKT and MAIT cells and how they operate during the course of chronic inflammatory diseases that eventually lead to hepatocellular carcinoma.
Collapse
|
39
|
Lee SK, Choi JY, Jung ES, Kwon JH, Jang JW, Bae SH, Yoon SK. An Immunological Perspective on the Mechanism of Drug Induced Liver Injury: Focused on Drugs for Treatment of Hepatocellular Carcinoma and Liver Transplantation. Int J Mol Sci 2023; 24:5002. [PMID: 36902432 PMCID: PMC10003078 DOI: 10.3390/ijms24055002] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
The liver is frequently exposed to potentially toxic materials, and it is the primary site of clearance of foreign agents, along with many innate and adaptive immune cells. Subsequently, drug induced liver injury (DILI), which is caused by medications, herbs, and dietary supplements, often occurs and has become an important issue in liver diseases. Reactive metabolites or drug-protein complexes induce DILI via the activation of various innate and adaptive immune cells. There has been a revolutionary development of treatment drugs for hepatocellular carcinoma (HCC) and liver transplantation (LT), including immune checkpoint inhibitors (ICIs), that show high efficacy in patients with advanced HCC. Along with the high efficacy of novel drugs, DILI has become a pivotal issue in the use of new drugs, including ICIs. This review demonstrates the immunological mechanism of DILI, including the innate and adaptive immune systems. Moreover, it aims to provide drug treatment targets, describe the mechanisms of DILI, and detail the management of DILI caused by drugs for HCC and LT.
Collapse
Affiliation(s)
- Soon Kyu Lee
- Division of Hepatology, Department of Internal Medicine, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jong Young Choi
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Hepatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Eun Sun Jung
- Department of Pathology, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jung Hyun Kwon
- Division of Hepatology, Department of Internal Medicine, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jeong Won Jang
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Hepatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Si Hyun Bae
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Hepatology, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Seung Kew Yoon
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Hepatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
40
|
Wiering L, Subramanian P, Hammerich L. Hepatic Stellate Cells: Dictating Outcome in Nonalcoholic Fatty Liver Disease. Cell Mol Gastroenterol Hepatol 2023; 15:1277-1292. [PMID: 36828280 PMCID: PMC10148161 DOI: 10.1016/j.jcmgh.2023.02.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 02/15/2023] [Accepted: 02/15/2023] [Indexed: 02/26/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a fast growing, chronic liver disease affecting ∼25% of the global population. Nonalcoholic fatty liver disease severity ranges from the less severe simple hepatic steatosis to the more advanced nonalcoholic steatohepatitis (NASH). The presence of NASH predisposes individuals to liver fibrosis, which can further progress to cirrhosis and hepatocellular carcinoma. This makes hepatic fibrosis an important indicator of clinical outcomes in patients with NASH. Hepatic stellate cell activation dictates fibrosis development during NASH. Here, we discuss recent advances in the analysis of the profibrogenic pathways and mediators of hepatic stellate cell activation and inactivation, which ultimately determine the course of disease in nonalcoholic fatty liver disease/NASH.
Collapse
Affiliation(s)
- Leke Wiering
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Junior Clinician Scientist Program, Berlin, Germany
| | - Pallavi Subramanian
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Linda Hammerich
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany.
| |
Collapse
|
41
|
Liu J, Ding M, Bai J, Luo R, Liu R, Qu J, Li X. Decoding the role of immune T cells: A new territory for improvement of metabolic-associated fatty liver disease. IMETA 2023; 2:e76. [PMID: 38868343 PMCID: PMC10989916 DOI: 10.1002/imt2.76] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/13/2022] [Accepted: 12/16/2022] [Indexed: 06/14/2024]
Abstract
Metabolic-associated fatty liver disease (MAFLD) is a new emerging concept and is associated with metabolic dysfunction, generally replacing the name of nonalcoholic fatty liver disease (NAFLD) due to heterogeneous liver condition and inaccuracies in definition. The prevalence of MAFLD is rising by year due to dietary changes, metabolic disorders, and no approved therapy, affecting a quarter of the global population and representing a major economic problem that burdens healthcare systems. Currently, in addition to the common causative factors like insulin resistance, oxidative stress, and lipotoxicity, the role of immune cells, especially T cells, played in MAFLD is increasingly being emphasized by global scholars. Based on the diverse classification and pathophysiological effects of immune T cells, we comprehensively analyzed their bidirectional regulatory effects on the hepatic inflammatory microenvironment and MAFLD progression. This interaction between MAFLD and T cells was also associated with hepatic-intestinal immune crosstalk and gut microbiota homeostasis. Moreover, we pointed out several T-cell-based therapeutic approaches including but not limited to adoptive transfer of T cells, fecal microbiota transplantation, and drug therapy, especially for natural products and Chinese herbal prescriptions. Overall, this study contributes to a better understanding of the important role of T cells played in MAFLD progression and corresponding therapeutic options and provides a potential reference for further drug development.
Collapse
Affiliation(s)
- Jia Liu
- School of Life SciencesBeijing University of Chinese MedicineBeijingChina
| | - Mingning Ding
- School of Life SciencesBeijing University of Chinese MedicineBeijingChina
| | - Jinzhao Bai
- School of Chinese Materia MedicaBeijing University of Chinese MedicineBeijingChina
| | - Ranyi Luo
- School of Life SciencesBeijing University of Chinese MedicineBeijingChina
| | - Runping Liu
- School of Chinese Materia MedicaBeijing University of Chinese MedicineBeijingChina
| | - Jiaorong Qu
- School of Life SciencesBeijing University of Chinese MedicineBeijingChina
| | - Xiaojiaoyang Li
- School of Life SciencesBeijing University of Chinese MedicineBeijingChina
| |
Collapse
|
42
|
Cannito S, Dianzani U, Parola M, Albano E, Sutti S. Inflammatory processes involved in NASH-related hepatocellular carcinoma. Biosci Rep 2023; 43:BSR20221271. [PMID: 36691794 PMCID: PMC9874450 DOI: 10.1042/bsr20221271] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/09/2022] [Accepted: 01/05/2023] [Indexed: 01/25/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the fourth leading cause of cancer-related death worldwide. In the recent years nonalcoholic fatty liver disease (NAFLD) is becoming a growing cause of HCCs and the incidence of NAFLD-related HCCs is expected to further dramatically increase by the next decade. Chronic inflammation is regarded as the driving force of NAFLD progression and a key factor in hepatic carcinogenesis. Hepatic inflammation in NAFLD results from the persistent stimulation of innate immunity in response to hepatocellular injury and gut dysbiosis as well as by the activation of adaptive immunity. However, the relative roles of innate and adaptive immunity in the processes leading to HCC are still incompletely characterized. This is due to the complex interplay between different liver cell populations, which is also strongly influenced by gut-derived bacterial products, metabolic/nutritional signals. Furthermore, carcinogenic mechanisms in NAFLD/NASH appear to involve the activation of signals mediated by hypoxia inducible factors. This review discusses recent data regarding the contribution of different inflammatory cells to NAFLD-related HCC and their possible impact on patient response to current treatments.
Collapse
Affiliation(s)
- Stefania Cannito
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Turin, Turin, Italy
| | - Umberto Dianzani
- Department of Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases, University of East Piedmont, Novara, Italy
| | - Maurizio Parola
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Turin, Turin, Italy
| | - Emanuele Albano
- Department of Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases, University of East Piedmont, Novara, Italy
| | - Salvatore Sutti
- Department of Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases, University of East Piedmont, Novara, Italy
| |
Collapse
|
43
|
Li Q, Chen F, Wang F. The immunological mechanisms and therapeutic potential in drug-induced liver injury: lessons learned from acetaminophen hepatotoxicity. Cell Biosci 2022; 12:187. [PMID: 36414987 PMCID: PMC9682794 DOI: 10.1186/s13578-022-00921-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 11/01/2022] [Indexed: 11/24/2022] Open
Abstract
Acute liver failure caused by drug overdose is a significant clinical problem in developed countries. Acetaminophen (APAP), a widely used analgesic and antipyretic drug, but its overdose can cause acute liver failure. In addition to APAP-induced direct hepatotoxicity, the intracellular signaling mechanisms of APAP-induced liver injury (AILI) including metabolic activation, mitochondrial oxidant stress and proinflammatory response further affect progression and severity of AILI. Liver inflammation is a result of multiple interactions of cell death molecules, immune cell-derived cytokines and chemokines, as well as damaged cell-released signals which orchestrate hepatic immune cell infiltration. The immunoregulatory interplay of these inflammatory mediators and switching of immune responses during AILI lead to different fate of liver pathology. Thus, better understanding the complex interplay of immune cell subsets in experimental models and defining their functional involvement in disease progression are essential to identify novel therapeutic targets for the treatment of AILI. Here, this present review aims to systematically elaborate on the underlying immunological mechanisms of AILI, its relevance to immune cells and their effector molecules, and briefly discuss great therapeutic potential based on inflammatory mediators.
Collapse
Affiliation(s)
- Qianhui Li
- grid.511083.e0000 0004 7671 2506Division of Gastroenterology, Seventh Affiliated Hospital of Sun Yat-sen University, No.628, Zhenyuan Road, Shenzhen, 518107 China
| | - Feng Chen
- grid.511083.e0000 0004 7671 2506Division of Gastroenterology, Seventh Affiliated Hospital of Sun Yat-sen University, No.628, Zhenyuan Road, Shenzhen, 518107 China
| | - Fei Wang
- grid.511083.e0000 0004 7671 2506Division of Gastroenterology, Seventh Affiliated Hospital of Sun Yat-sen University, No.628, Zhenyuan Road, Shenzhen, 518107 China
| |
Collapse
|
44
|
Hu H, Sun N, Du H, He Y, Pan K, Liu X, Lu X, Wei J, Liao M, Duan C. Mouse promyelocytic leukemia zinc finger protein (PLZF) regulates hepatic lipid and glucose homeostasis dependent on SIRT1. Front Pharmacol 2022; 13:1039726. [PMID: 36438786 PMCID: PMC9684722 DOI: 10.3389/fphar.2022.1039726] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 10/21/2022] [Indexed: 11/11/2022] Open
Abstract
Previous studies have demonstrated that promyelocytic leukemia zinc finger protein (PLZF) promotes the expression of gluconeogenic genes and hepatic glucose output, which leads to hyperglycemia. However, the role played by PLZF in regulating lipid metabolism is not known. In this study, we aimed to examine the function of PLZF in regulating hepatic lipid and glucose homeostasis and the underlying mechanisms. The expression of PLZF was determined in different mouse models with regard to non-alcoholic fatty liver disease (NAFLD). In the next step, adenoviruses that express PLZF (Ad-PLZF) or PLZF-specific shRNA (Ad-shPLZF) were utilized to alter PLZF expression in mouse livers and in primary hepatocytes. For the phenotype of the fatty liver, histologic and biochemical analyses of hepatic triglyceride (TG), serum TG and cholesterol levels were carried out. The underlying molecular mechanism for the regulation of lipid metabolism by PLZF was further explored using luciferase reporter gene assay and ChIP analysis. The results demonstrated that PLZF expression was upregulated in livers derived from ob/ob, db/db and diet-induced obesity (DIO) mice. Liver PLZF-overexpressing C57BL/6J mice showed fatty liver phenotype, liver inflammation, impaired glucose tolerance and insulin sensitivity. On the other hand, hepatic PLZF knockdown in db/db and DIO mice alleviated hepatic steatosis. Of note, we found that PLZF activates SREBP-1c gene transcription through binding directly to the promoter fragment of this gene, which would induce a repressor-to-activator conversion depending on its interaction with SIRT1 in the role played by PLZF in the transcription process through deacetylation. Thus, PLZF is identified as an essential regulator of hepatic lipid and glucose metabolism, where the modulation of its liver expression could open up a therapeutic path for treating NAFLD.
Collapse
Affiliation(s)
- Huiling Hu
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Nannan Sun
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Haiyan Du
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Yuqing He
- Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Kunyi Pan
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiuli Liu
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiaoxia Lu
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jie Wei
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- *Correspondence: Jie Wei, ; Mianmian Liao, ; Chaohui Duan,
| | - Mianmian Liao
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- *Correspondence: Jie Wei, ; Mianmian Liao, ; Chaohui Duan,
| | - Chaohui Duan
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- *Correspondence: Jie Wei, ; Mianmian Liao, ; Chaohui Duan,
| |
Collapse
|
45
|
Yu W, Li C, Zhang D, Li Z, Xia P, Liu X, Cai X, Yang P, Ling J, Zhang J, Zhang M, Yu P. Advances in T Cells Based on Inflammation in Metabolic Diseases. Cells 2022; 11:cells11223554. [PMID: 36428983 PMCID: PMC9688178 DOI: 10.3390/cells11223554] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 10/26/2022] [Accepted: 11/02/2022] [Indexed: 11/12/2022] Open
Abstract
With the increasing incidence of metabolic diseases year by year and their impact on the incidence of cardiovascular diseases, metabolic diseases have attracted great attention as a major health care problem, but there is still no effective treatment. Oxidative stress and inflammation are the main mechanisms leading to metabolic diseases. T cells are involved in the inflammatory response, which can also regulate the development of metabolic diseases, CD4+ T cells and CD8+ T cells are mainly responsible for the role. Th1 and Th17 differentiated from CD4+ T promote inflammation, while Th2 and Treg inhibit inflammation. CD8+ T cells also contribute to inflammation. The severity and duration of inflammatory reactions can also lead to different degrees of progression of metabolic diseases. Moreover, mTOR, PI3K-Akt, and AMPK signaling pathways play unique roles in the regulation of T cells, which provide a new direction for the treatment of metabolic diseases in the future. In this review, we will elaborate on the role of T cells in regulating inflammation in various metabolic diseases, the signaling pathways that regulate T cells in metabolic diseases, and the latest research progress.
Collapse
Affiliation(s)
- Wenlu Yu
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, China
- School of Ophthalmology and Optometry, Nanchang University, Nanchang 330000, China
| | - Chunxiu Li
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, China
- School of Ophthalmology and Optometry, Nanchang University, Nanchang 330000, China
| | - Deju Zhang
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong 999077, China
| | - Zhangwang Li
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, China
- The Second Clinical Medical College, Nanchang University, Nanchang 330000, China
| | - Panpan Xia
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, China
| | - Xiao Liu
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510000, China
| | - Xia Cai
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, China
| | - Pingping Yang
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, China
| | - Jitao Ling
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, China
- Correspondence: (J.Z.); (P.Y.)
| | - Meiying Zhang
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, China
| | - Peng Yu
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, China
- Correspondence: (J.Z.); (P.Y.)
| |
Collapse
|
46
|
Koda Y, Nakamoto N, Kanai T. Regulation of Progression and Resolution of Liver Fibrosis by Immune Cells. Semin Liver Dis 2022; 42:475-488. [PMID: 36208620 DOI: 10.1055/a-1957-6384] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The excessive accumulation of extracellular matrix proteins results in fibrosis-a condition implicated in several diseased conditions, such as nonalcoholic steatohepatitis, viral hepatitis, and autoimmune hepatitis. Despite its prevalence, direct and effective treatments for fibrosis are lacking, warranting the development of better therapeutic strategies. Accumulating evidence has shown that liver fibrosis-a condition previously considered irreversible-is reversible in specific conditions. Immune cells residing in or infiltrating the liver (e.g., macrophages) are crucial in the pathogenesis of fibrosis. Given this background, the roles and action mechanisms of various immune cells and their subsets in the progression and recovery of liver fibrosis, particularly concerning nonalcoholic steatohepatitis, are discussed in this review. Furthermore, the development of better therapeutic strategies based on stage-specific properties and using advanced techniques as well as the mechanisms underlying recovery are elaborated. In conclusion, we consider the review comprehensively provides the present achievements and future possibilities revolving around fibrosis treatment.
Collapse
Affiliation(s)
- Yuzo Koda
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan.,Mitsubishi Tanabe Pharma Corporation, Kanagawa, Japan
| | - Nobuhiro Nakamoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan.,Japan Agency for Medical Research and Development, Japan Agency for Medical Research and Development, Tokyo, Japan
| |
Collapse
|
47
|
Wang SX, Yan JS, Chan YS. Advancements in MAFLD Modeling with Human Cell and Organoid Models. Int J Mol Sci 2022; 23:11850. [PMID: 36233151 PMCID: PMC9569457 DOI: 10.3390/ijms231911850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/17/2022] Open
Abstract
Metabolic (dysfunction) associated fatty liver disease (MAFLD) is one of the most prevalent liver diseases and has no approved therapeutics. The high failure rates witnessed in late-phase MAFLD drug trials reflect the complexity of the disease, and how the disease develops and progresses remains to be fully understood. In vitro, human disease models play a pivotal role in mechanistic studies to unravel novel disease drivers and in drug testing studies to evaluate human-specific responses. This review focuses on MAFLD disease modeling using human cell and organoid models. The spectrum of patient-derived primary cells and immortalized cell lines employed to model various liver parenchymal and non-parenchymal cell types essential for MAFLD development and progression is discussed. Diverse forms of cell culture platforms utilized to recapitulate tissue-level pathophysiology in different stages of the disease are also reviewed.
Collapse
Affiliation(s)
- Shi-Xiang Wang
- Guangzhou Laboratory, No. 9 Xing Dao Huan Bei Road, Guangzhou International Bio Island, Guangzhou 510005, China
| | - Ji-Song Yan
- Guangzhou Laboratory, No. 9 Xing Dao Huan Bei Road, Guangzhou International Bio Island, Guangzhou 510005, China
- School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Yun-Shen Chan
- Guangzhou Laboratory, No. 9 Xing Dao Huan Bei Road, Guangzhou International Bio Island, Guangzhou 510005, China
| |
Collapse
|
48
|
Immune mechanisms linking metabolic injury to inflammation and fibrosis in fatty liver disease - novel insights into cellular communication circuits. J Hepatol 2022; 77:1136-1160. [PMID: 35750137 DOI: 10.1016/j.jhep.2022.06.012] [Citation(s) in RCA: 263] [Impact Index Per Article: 87.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 06/02/2022] [Accepted: 06/06/2022] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver disease and is emerging as the leading cause of cirrhosis, liver transplantation and hepatocellular carcinoma (HCC). NAFLD is a metabolic disease that is considered the hepatic manifestation of the metabolic syndrome; however, during the evolution of NAFLD from steatosis to non-alcoholic steatohepatitis (NASH), to more advanced stages of NASH with liver fibrosis, the immune system plays an integral role. Triggers for inflammation are rooted in hepatic (lipid overload, lipotoxicity, oxidative stress) and extrahepatic (gut-liver axis, adipose tissue, skeletal muscle) systems, resulting in unique immune-mediated pathomechanisms in NAFLD. In recent years, the implementation of single-cell RNA-sequencing and high dimensional multi-omics (proteogenomics, lipidomics) and spatial transcriptomics have tremendously advanced our understanding of the complex heterogeneity of various liver immune cell subsets in health and disease. In NAFLD, several emerging inflammatory mechanisms have been uncovered, including profound macrophage heterogeneity, auto-aggressive T cells, the role of unconventional T cells and platelet-immune cell interactions, potentially yielding novel therapeutics. In this review, we will highlight the recent discoveries related to inflammation in NAFLD, discuss the role of immune cell subsets during the different stages of the disease (including disease regression) and integrate the multiple systems driving inflammation. We propose a refined concept by which the immune system contributes to all stages of NAFLD and discuss open scientific questions arising from this paradigm shift that need to be unravelled in the coming years. Finally, we discuss novel therapeutic approaches to target the multiple triggers of inflammation, including combination therapy via nuclear receptors (FXR agonists, PPAR agonists).
Collapse
|
49
|
Hoogerland JA, Staels B, Dombrowicz D. Immune-metabolic interactions in homeostasis and the progression to NASH. Trends Endocrinol Metab 2022; 33:690-709. [PMID: 35961913 DOI: 10.1016/j.tem.2022.07.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 07/09/2022] [Accepted: 07/12/2022] [Indexed: 12/16/2022]
Abstract
The incidence of non-alcoholic fatty liver disease (NAFLD) has increased significantly over the past two decades. NAFLD ranges from simple steatosis (NAFL) to nonalcoholic steatohepatitis (NASH) and predisposes to fibrosis and hepatocellular carcinoma (HCC). The importance of the immune system in hepatic physiology and in the progression of NAFLD is increasingly recognized. At homeostasis, the liver participates in immune defense against pathogens and in tolerance of gut-derived microbial compounds. Hepatic immune cells also respond to metabolic stimuli and have a role in NAFLD progression to NASH. In this review, we discuss how metabolic perturbations affect immune cell phenotype and function in NAFL and NASH, and then focus on the role of immune cells in liver homeostasis and in the development of NASH.
Collapse
Affiliation(s)
- Joanne A Hoogerland
- Univeristy of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Bart Staels
- Univeristy of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - David Dombrowicz
- Univeristy of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France.
| |
Collapse
|
50
|
Zhang P, Chen Z, Kuang H, Liu T, Zhu J, Zhou L, Wang Q, Xiong X, Meng Z, Qiu X, Jacks R, Liu L, Li S, Lumeng CN, Li Q, Zhou X, Lin JD. Neuregulin 4 suppresses NASH-HCC development by restraining tumor-prone liver microenvironment. Cell Metab 2022; 34:1359-1376.e7. [PMID: 35973424 PMCID: PMC9458631 DOI: 10.1016/j.cmet.2022.07.010] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 04/20/2022] [Accepted: 07/20/2022] [Indexed: 12/13/2022]
Abstract
The mammalian liver comprises heterogeneous cell types within its tissue microenvironment that undergo pathophysiological reprogramming in disease states, such as non-alcoholic steatohepatitis (NASH). Patients with NASH are at an increased risk for the development of hepatocellular carcinoma (HCC). However, the molecular and cellular nature of liver microenvironment remodeling that links NASH to liver carcinogenesis remains obscure. Here, we show that diet-induced NASH is characterized by the induction of tumor-associated macrophage (TAM)-like macrophages and exhaustion of cytotoxic CD8+ T cells in the liver. The adipocyte-derived endocrine factor Neuregulin 4 (NRG4) serves as a hormonal checkpoint that restrains this pathological reprogramming during NASH. NRG4 deficiency exacerbated the induction of tumor-prone liver immune microenvironment and NASH-related HCC, whereas transgenic NRG4 overexpression elicited protective effects in mice. In a therapeutic setting, recombinant NRG4-Fc fusion protein exhibited remarkable potency in suppressing HCC and prolonged survival in the treated mice. These findings pave the way for therapeutic intervention of liver cancer by targeting the NRG4 hormonal checkpoint.
Collapse
Affiliation(s)
- Peng Zhang
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Zhimin Chen
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Henry Kuang
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Tongyu Liu
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Jiaqiang Zhu
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Linkang Zhou
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Qiuyu Wang
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Xuelian Xiong
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Ziyi Meng
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Xiaoxue Qiu
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Ramiah Jacks
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Lu Liu
- Department of Internal Medicine and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Siming Li
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Carey N Lumeng
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Qing Li
- Department of Internal Medicine and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Xiang Zhou
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA; Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jiandie D Lin
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA.
| |
Collapse
|