1
|
Liu Y, Wang TT, Lu Y, Riaz M, Qyang Y. Cardiac macrophage: Insights from murine models to translational potential for human studies. J Mol Cell Cardiol 2025; 204:17-31. [PMID: 40354877 DOI: 10.1016/j.yjmcc.2025.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
Macrophages are a cell type that are known to play dynamic roles in acute and progressive pathology. They are highly attuned to their microenvironments throughout maturation, tailoring their functional responses according to the specific tissues in which they reside and their developmental origin. Cardiac macrophages (cMacs) have emerged as focal points of interest for their interactions with the unique electrical and mechanical stimuli of the heart, as well as for their role in maintaining cardiac homeostasis. Through an in-depth analysis of their origin, lineage, and functional significance, this review aims to shed light on cMacs' distinct contributions to both normal physiological maintenance as well as disease progression. Central to our discussion is the comparison of cMac characteristics between mouse and human models, highlighting current challenges and proposing novel experimental tools for deciphering cMac function within the intricate human cardiac microenvironments based on current murine studies. Our review offers valuable insights for identifying novel therapeutic targets and interventions tailored to the distinct roles of these immune cells in cardiovascular diseases (CVDs).
Collapse
Affiliation(s)
- Yufeng Liu
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department, New Haven, CT, USA; Yale Biological and Biomedical Sciences, Graduate School of Arts and Sciences, Yale University, New Haven, CT, USA
| | - Tricia T Wang
- Yale Biological and Biomedical Sciences, Graduate School of Arts and Sciences, Yale University, New Haven, CT, USA
| | - Yinsheng Lu
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department, New Haven, CT, USA; Yale Stem Cell Center, New Haven, CT, USA; Department of Mechanical Engineering and Materials Science, Graduate School of Arts and Sciences, Yale University, New Haven, CT, USA
| | - Muhammad Riaz
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department, New Haven, CT, USA; Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Yibing Qyang
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department, New Haven, CT, USA; Department of Pathology, Yale School of Medicine, New Haven, CT, USA; Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
| |
Collapse
|
2
|
Chiu DKC, Zhang X, Cheng BYL, Liu Q, Hayashi K, Yu B, Lee R, Zhang C, An X, Rajadas J, Reticker-Flynn NE, Rankin EB, Engleman EG. Tumor-derived erythropoietin acts as an immunosuppressive switch in cancer immunity. Science 2025; 388:eadr3026. [PMID: 40273234 PMCID: PMC12110762 DOI: 10.1126/science.adr3026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 12/20/2024] [Accepted: 03/05/2025] [Indexed: 04/26/2025]
Abstract
Successful cancer immunotherapy requires a patient to mount an effective immune response against tumors; however, many cancers evade the body's immune system. To investigate the basis for treatment failure, we examined spontaneous mouse models of hepatocellular carcinoma (HCC) with either an inflamed T cell-rich or a noninflamed T cell-deprived tumor microenvironment (TME). Our studies reveal that erythropoietin (EPO) secreted by tumor cells determines tumor immunotype. Tumor-derived EPO autonomously generates a noninflamed TME by interacting with its cognate receptor EPOR on tumor-associated macrophages (TAMs). EPO signaling prompts TAMs to become immunoregulatory through NRF2-mediated heme depletion. Removing either tumor-derived EPO or EPOR on TAMs leads to an inflamed TME and tumor regression independent of genotype, owing to augmented antitumor T cell immunity. Thus, the EPO/EPOR axis functions as an immunosuppressive switch for antitumor immunity.
Collapse
Affiliation(s)
| | - Xiangyue Zhang
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | | | - Qiang Liu
- Biomaterials and Advanced Drug Delivery Laboratory, Stanford University School of Medicine, Stanford, CA, USA
| | - Kazukuni Hayashi
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Bo Yu
- ImmunEdge Inc. Mountain View, California 94043, USA
| | - Ryan Lee
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Catherine Zhang
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Xiuli An
- Laboratory of Membrane Biology, New York Blood Center, New York, NY, 10065, USA
| | - Jayakumar Rajadas
- Biomaterials and Advanced Drug Delivery Laboratory, Stanford University School of Medicine, Stanford, CA, USA
| | - Nathan E Reticker-Flynn
- Department of Otolaryngology - Head and Neck Surgery, Stanford University, Stanford, CA 94305, USA
| | - Erinn B Rankin
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
- Stanford Cancer Institute, Stanford University, Palo Alto, CA 94305, USA
| | - Edgar G Engleman
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
- Stanford Cancer Institute, Stanford University, Palo Alto, CA 94305, USA
- Lead contact
| |
Collapse
|
3
|
Pessenda G, Ferreira TR, Paun A, Kabat J, Amaral EP, Kamenyeva O, Gazzinelli-Guimaraes PH, Perera SR, Ganesan S, Lee SH, Sacks DL. Kupffer cell and recruited macrophage heterogeneity orchestrate granuloma maturation and hepatic immunity in visceral leishmaniasis. Nat Commun 2025; 16:3125. [PMID: 40169598 PMCID: PMC11961706 DOI: 10.1038/s41467-025-58360-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 03/19/2025] [Indexed: 04/03/2025] Open
Abstract
In murine models of visceral leishmaniasis (VL), the parasitization of resident Kupffer cells (resKCs) drives early Leishmania infantum growth in the liver, leading to granuloma formation and subsequent parasite control. Using the chronic VL model, we demonstrate that polyclonal resKCs redistributed to form granulomas outside the sinusoids, creating an open sinusoidal niche that was gradually repopulated by monocyte-derived KCs (moKCs) acquiring a tissue specific, homeostatic profile. Early-stage granulomas predominantly consisted of CLEC4F+KCs. In contrast, late-stage granulomas led to remodeling of the sinusoidal network and contained monocyte-derived macrophages (momacs) along with KCs that downregulated CLEC4F, with both populations expressing iNOS and pro-inflammatory chemokines. During late-stage infection, parasites were largely confined to CLEC4F-KCs. Reduced monocyte recruitment and increased resKCs proliferation in infected Ccr2-/- mice impaired parasite control. These findings show that the ontogenic heterogeneity of granuloma macrophages is closely linked to granuloma maturation and the development of hepatic immunity in VL.
Collapse
Affiliation(s)
- Gabriela Pessenda
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Tiago R Ferreira
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Andrea Paun
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Juraj Kabat
- Biological Imaging Section, Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Eduardo P Amaral
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Olena Kamenyeva
- Biological Imaging Section, Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Pedro Henrique Gazzinelli-Guimaraes
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Department of Microbiology, Immunology & Tropical Medicine School of Medicine & Health Sciences. The George Washington University, Washington DC, USA
| | - Shehan R Perera
- Department of Electrical and Computer Engineering, The Ohio State University, Columbus, OH, USA
| | - Sundar Ganesan
- Biological Imaging Section, Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sang Hun Lee
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - David L Sacks
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
4
|
Ramirez CFA, Akkari L. Myeloid cell path to malignancy: insights into liver cancer. Trends Cancer 2025:S2405-8033(25)00054-8. [PMID: 40140328 DOI: 10.1016/j.trecan.2025.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/12/2025] [Accepted: 02/24/2025] [Indexed: 03/28/2025]
Abstract
Clinically approved treatments for advanced liver cancer often lack potency because of the heterogeneous characteristics of hepatocellular carcinoma (HCC). This complexity is largely driven by context-dependent inflammatory responses brought on by diverse etiologies, such as metabolic dysfunction-associated steatohepatitis (MASH), the genetic makeup of cancer cells, and the versatile adaptability of immune cells, such as myeloid cells. In this review, we discuss the evolutionary dynamics of the immune landscape, particularly that of liver-resident Kupffer cells (KCs), TREM2+, and SPP1+ macrophages with an active role during liver disease progression, which eventually fuels hepatocarcinogenesis. We highlight exploitable immunomodulatory avenues amenable to mitigate both the inherent pathological characteristics of liver cancers and the associated external factors that favor malignancy, paving a roadmap toward improving the management and therapeutic outcome for patients with HCC.
Collapse
Affiliation(s)
- Christel F A Ramirez
- Division of Tumor Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Leila Akkari
- Division of Tumor Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands.
| |
Collapse
|
5
|
Ramachandran P. Zonal control of liver inflammation is orchestrated by periportal Kupffer cells. J Hepatol 2025; 82:546-547. [PMID: 39824734 DOI: 10.1016/j.jhep.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 01/20/2025]
Affiliation(s)
- Prakash Ramachandran
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, United Kingdom.
| |
Collapse
|
6
|
Vishnyakova P, Elchaninov A, Fatkhudinov T, Kolesov D. Unravelling approaches to study macrophages: from classical to novel biophysical methodologies. PeerJ 2025; 13:e19039. [PMID: 39989743 PMCID: PMC11847493 DOI: 10.7717/peerj.19039] [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: 10/03/2024] [Accepted: 01/31/2025] [Indexed: 02/25/2025] Open
Abstract
Macrophages play crucial roles in immune responses and tissue homeostasis. Despite the fact that macrophages were described more than a century ago, they continue to be the cells of intensive interest. Advanced understanding of phenotypic diversity in macrophages holds great promise for development of cell-based therapeutic strategies. The introduction of innovative approaches in cell biology greatly enhances our ability to investigate the unique characteristics of macrophages. The review considers both classical methods to study macrophages and high-tech approaches, including single-cell sequencing, single-cell mass spectrometry, droplet microfluidics, scanning probe microscopy and atomic force spectroscopy. This review will be valuable both to specialists beginning their study of macrophages and to experienced scientists seeking to deepen their understanding of methods at the intersection of biological and physical sciences.
Collapse
Affiliation(s)
- Polina Vishnyakova
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V. I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), Moscow, Russia
| | - Andrey Elchaninov
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V. I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), Moscow, Russia
- A.P. Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, Moscow, Russia
| | - Timur Fatkhudinov
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), Moscow, Russia
- A.P. Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, Moscow, Russia
| | - Dmitry Kolesov
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V. I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
- Moscow Polytechnic University, Moscow, Russia
| |
Collapse
|
7
|
Ma X, Huang T, Chen X, Li Q, Liao M, Fu L, Huang J, Yuan K, Wang Z, Zeng Y. Molecular mechanisms in liver repair and regeneration: from physiology to therapeutics. Signal Transduct Target Ther 2025; 10:63. [PMID: 39920130 PMCID: PMC11806117 DOI: 10.1038/s41392-024-02104-8] [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: 11/08/2023] [Revised: 09/02/2024] [Accepted: 12/12/2024] [Indexed: 02/09/2025] Open
Abstract
Liver repair and regeneration are crucial physiological responses to hepatic injury and are orchestrated through intricate cellular and molecular networks. This review systematically delineates advancements in the field, emphasizing the essential roles played by diverse liver cell types. Their coordinated actions, supported by complex crosstalk within the liver microenvironment, are pivotal to enhancing regenerative outcomes. Recent molecular investigations have elucidated key signaling pathways involved in liver injury and regeneration. Viewed through the lens of metabolic reprogramming, these pathways highlight how shifts in glucose, lipid, and amino acid metabolism support the cellular functions essential for liver repair and regeneration. An analysis of regenerative variability across pathological states reveals how disease conditions influence these dynamics, guiding the development of novel therapeutic strategies and advanced techniques to enhance liver repair and regeneration. Bridging laboratory findings with practical applications, recent clinical trials highlight the potential of optimizing liver regeneration strategies. These trials offer valuable insights into the effectiveness of novel therapies and underscore significant progress in translational research. In conclusion, this review intricately links molecular insights to therapeutic frontiers, systematically charting the trajectory from fundamental physiological mechanisms to innovative clinical applications in liver repair and regeneration.
Collapse
Affiliation(s)
- Xiao Ma
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Tengda Huang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xiangzheng Chen
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qian Li
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Mingheng Liao
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Li Fu
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jiwei Huang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Kefei Yuan
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Zhen Wang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| | - Yong Zeng
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
8
|
Frenis K, Badalamenti B, Mamigonian O, Weng C, Wang D, Fierstein S, Côté P, Khong H, Li H, Lummertz da Rocha E, Sankaran VG, Rowe RG. Path of differentiation defines human macrophage identity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.24.634694. [PMID: 39896569 PMCID: PMC11785145 DOI: 10.1101/2025.01.24.634694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Macrophages play central roles in immunity, wound healing, and homeostasis - a functional diversity that is underpinned by varying developmental origins. The impact of ontogeny on properties of human macrophages is inadequately understood. We demonstrate that definitive human fetal liver (HFL) hematopoietic stem cells (HSCs) possess two divergent paths of macrophage specification that lead to distinct identities. The monocyte-dependent pathway exists in both prenatal and postnatal hematopoiesis and generates macrophages with adult-like responses properties. We now uncover a fetal-specific pathway of expedited differentiation that generates tissue resident-like macrophages (TRMs) that retain HSC-like self-renewal programs governed by the aryl hydrocarbon receptor (AHR). We show that AHR antagonism promotes TRM expansion and mitigates inflammation in models of atopic dermatitis (AD). Overall, we directly connect path of differentiation with functional properties of macrophages and identify an approach to promote selective expansion of TRMs with direct relevance to inflammation and diseases of macrophage dysfunction.
Collapse
|
9
|
Yan M, Cui Y, Xiang Q. Metabolism of hepatic stellate cells in chronic liver diseases: emerging molecular and therapeutic interventions. Theranostics 2025; 15:1715-1740. [PMID: 39897543 PMCID: PMC11780521 DOI: 10.7150/thno.106597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 12/10/2024] [Indexed: 02/04/2025] Open
Abstract
Chronic liver diseases, primarily metabolic dysfunction-associated steatotic liver disease (MASLD), metabolic and metabolic dysfunction-associated alcoholic liver disease (MetALD), and viral hepatitis, can lead to liver fibrosis, cirrhosis, and cancer. Hepatic stellate cell (HSC) activation plays a central role in the development of myofibroblasts and fibrogenesis in chronic liver diseases. However, HSC activation is influenced by the complex microenvironments within the liver, which are largely shaped by the interactions between HSCs and various other cell types. Changes in HSC phenotypes and metabolic mechanisms involve glucose, lipid, and cholesterol metabolism, oxidative stress, activation of the unfolded protein response (UPR), autophagy, ferroptosis, senescence, and nuclear receptors. Clinical interventions targeting these pathways have shown promising results in addressing liver inflammation and fibrosis, as well as in modulating glucose and lipid metabolism and metabolic stress responses. Therefore, a comprehensive understanding of HSC phenotypes and metabolic mechanisms presents opportunities for novel therapeutic approaches aimed at halting or even reversing chronic liver diseases.
Collapse
Affiliation(s)
- Mengyao Yan
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, China
| | - Yimin Cui
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, China
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - Qian Xiang
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, China
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| |
Collapse
|
10
|
Ran J, Yin S, Issa R, Zhao Q, Zhu G, Zhang H, Zhang Q, Wu C, Li J. Key role of macrophages in the progression of hepatic fibrosis. Hepatol Commun 2025; 9:e0602. [PMID: 39670853 PMCID: PMC11637753 DOI: 10.1097/hc9.0000000000000602] [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: 05/08/2024] [Accepted: 09/10/2024] [Indexed: 12/14/2024] Open
Abstract
Liver fibrosis is a pathological change characterized by excessive deposition of extracellular matrix caused by chronic liver injury, and the mechanisms underlying its development are associated with endothelial cell injury, inflammatory immune cell activation, and HSC activation. Furthermore, hepatic macrophages exhibit remarkable heterogeneity and hold central functions in the evolution of liver fibrosis, with different subgroups exerting dual effects of promotion and regression. Currently, targeted macrophage therapy for reversing hepatic fibrosis has been extensively studied and has shown promising prospects. In this review, we will discuss the dual role of macrophages in liver fibrosis and provide new insights into reversing liver fibrosis based on macrophages.
Collapse
Affiliation(s)
- Jinqiu Ran
- Department of Infectious Disease, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
- Department of Infectious Disease, Institute of Viruses and Infectious Diseases, Nanjing University, Nanjing, Jiangsu, China
| | - Shengxia Yin
- Department of Infectious Disease, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
- Department of Infectious Disease, Institute of Viruses and Infectious Diseases, Nanjing University, Nanjing, Jiangsu, China
| | - Rahma Issa
- Department of Infectious Disease, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
- Department of Infectious Disease, Institute of Viruses and Infectious Diseases, Nanjing University, Nanjing, Jiangsu, China
| | - Qianwen Zhao
- Department of Infectious Disease, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
- Department of Infectious Disease, Institute of Viruses and Infectious Diseases, Nanjing University, Nanjing, Jiangsu, China
| | - Guangqi Zhu
- Department of Infectious Disease, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
- Department of Infectious Disease, Institute of Viruses and Infectious Diseases, Nanjing University, Nanjing, Jiangsu, China
| | - Huan Zhang
- Department of Infectious Diseases, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Qun Zhang
- Department of Infectious Diseases, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Chao Wu
- Department of Infectious Disease, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
- Department of Infectious Disease, Institute of Viruses and Infectious Diseases, Nanjing University, Nanjing, Jiangsu, China
| | - Jie Li
- Department of Infectious Disease, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
- Department of Infectious Disease, Institute of Viruses and Infectious Diseases, Nanjing University, Nanjing, Jiangsu, China
| |
Collapse
|
11
|
Musrati MA, Stijlemans B, Azouz A, Kancheva D, Mesbahi S, Hadadi E, Lebegge E, Ali L, De Vlaminck K, Scheyltjens I, Vandamme N, Zivalj M, Assaf N, Elkrim Y, Ahmidi I, Huart C, Lamkanfi M, Guilliams M, De Baetselier P, Goriely S, Movahedi K, Van Ginderachter JA. Infection history imprints prolonged changes to the epigenome, transcriptome and function of Kupffer cells. J Hepatol 2024; 81:1023-1039. [PMID: 39002639 DOI: 10.1016/j.jhep.2024.07.007] [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: 08/03/2023] [Revised: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND & AIMS Liver macrophages fulfill various homeostatic functions and represent an essential line of defense against pathogenic insults. However, it remains unclear whether a history of infectious disease in the liver leads to long-term alterations to the liver macrophage compartment. METHODS We utilized a curable model of parasitic infection invoked by the protozoan parasite Trypanosoma brucei brucei to investigate whether infection history can durably reshape hepatic macrophage identity and function. Employing a combination of fate mapping, single-cell CITE-sequencing, single-nuclei multiome analysis, epigenomic analysis, and functional assays, we studied the alterations to the liver macrophage compartment during and after the resolution of infection. RESULTS We show that T. brucei brucei infection alters the composition of liver-resident macrophages, leading to the infiltration of monocytes that differentiate into various infection-associated macrophage populations with divergent transcriptomic profiles. Whereas infection-associated macrophages disappear post-resolution of infection, monocyte-derived macrophages engraft in the liver, assume a Kupffer cell (KC)-like profile and co-exist with embryonic KCs in the long-term. Remarkably, the prior exposure to infection imprinted an altered transcriptional program on post-resolution KCs that was underpinned by an epigenetic remodeling of KC chromatin landscapes and a shift in KC ontogeny, along with transcriptional and epigenetic alterations in their niche cells. This reprogramming altered KC functions and was associated with increased resilience to a subsequent bacterial infection. CONCLUSION Our study demonstrates that a prior exposure to a parasitic infection induces trained immunity in KCs, reshaping their identity and function in the long-term. IMPACT AND IMPLICATIONS Although the liver is frequently affected during infections, and despite housing a major population of resident macrophages known as Kupffer cells (KCs), it is currently unclear whether infections can durably alter KCs and their niche cells. Our study provides a comprehensive investigation into the long-term impact of a prior, cured parasitic infection, unveiling long-lasting ontogenic, epigenetic, transcriptomic and functional changes to KCs as well as KC niche cells, which may contribute to KC remodeling. Our data suggest that infection history may continuously reprogram KCs throughout life with potential implications for subsequent disease susceptibility in the liver, influencing preventive and therapeutic approaches.
Collapse
Affiliation(s)
- Mohamed Amer Musrati
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium; Cellular and Molecular Immunology Lab, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium
| | - Benoit Stijlemans
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium; Cellular and Molecular Immunology Lab, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium
| | - Abdulkader Azouz
- Institute for Medical Immunology, Université Libre de Bruxelles (ULB), Gosselies, Belgium; ULB Center for Research in Immunology (U-CRI), Gosselies, Belgium
| | - Daliya Kancheva
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium; Cellular and Molecular Immunology Lab, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium; Brain and Systems Immunology Lab, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium
| | - Sarah Mesbahi
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium; Cellular and Molecular Immunology Lab, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium
| | - Eva Hadadi
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium; Cellular and Molecular Immunology Lab, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium
| | - Els Lebegge
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium; Cellular and Molecular Immunology Lab, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium
| | - Leen Ali
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium; Cellular and Molecular Immunology Lab, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium; Brain and Systems Immunology Lab, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium
| | - Karen De Vlaminck
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium; Cellular and Molecular Immunology Lab, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium; Brain and Systems Immunology Lab, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium
| | - Isabelle Scheyltjens
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium; Cellular and Molecular Immunology Lab, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium; Brain and Systems Immunology Lab, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium
| | - Niels Vandamme
- Data Mining and Modeling for Biomedicine, VIB-UGent Center for Inflammation Research, Ghent, Belgium; VIB Single Cell Core, VIB, Ghent-Leuven, Belgium
| | - Maida Zivalj
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium; Cellular and Molecular Immunology Lab, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium
| | - Naela Assaf
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium; Cellular and Molecular Immunology Lab, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium
| | - Yvon Elkrim
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium; Cellular and Molecular Immunology Lab, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium
| | - Ilham Ahmidi
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium; Cellular and Molecular Immunology Lab, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium
| | - Camille Huart
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium; Cellular and Molecular Immunology Lab, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium
| | - Mohamed Lamkanfi
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Martin Guilliams
- Laboratory of Myeloid Cell Biology in Tissue Homeostasis and Regeneration, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Faculty of Sciences, Ghent University, Ghent, Belgium
| | - Patrick De Baetselier
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium; Cellular and Molecular Immunology Lab, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium
| | - Stanislas Goriely
- Institute for Medical Immunology, Université Libre de Bruxelles (ULB), Gosselies, Belgium; ULB Center for Research in Immunology (U-CRI), Gosselies, Belgium
| | - Kiavash Movahedi
- Brain and Systems Immunology Lab, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium.
| | - Jo A Van Ginderachter
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium; Cellular and Molecular Immunology Lab, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
12
|
Schwenger KJP, Ghorbani Y, Bharatselvam S, Chen L, Chomiak KM, Tyler AC, Eddingsaas NC, Fischer SE, Jackson TD, Okrainec A, Allard JP. Links between fecal microplastics and parameters related to metabolic dysfunction-associated steatotic liver disease (MASLD) in humans: An exploratory study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 953:176153. [PMID: 39260480 DOI: 10.1016/j.scitotenv.2024.176153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/06/2024] [Accepted: 09/07/2024] [Indexed: 09/13/2024]
Abstract
Microplastics (MPs) can persist in the environment and human body. Murine studies showed that exposure to MPs could cause metabolic dysregulation, contributing metabolic dysfunction-associated steatotic liver disease (MASLD) or steatohepatitis (MASH). However, research on the role of MPs in humans is limited. Thus, we aimed to assess links between human fecal MPs and liver histology, gene expression, immune cells and intestinal microbiota (IM). We included 6 lean healthy liver donors and 6 normal liver (obese) and 11 MASH patients. Overall, pre-BSx, we observed no significant differences in fecal MPs between groups. However, fecal MP fibers and total MPs positively correlated with portal and total macrophages and total killer T cells while total fecal MPs were positively correlated with natural killer cells. Additionally, 19 genes related to immune system and apoptosis correlated with fecal MPs at baseline. Fecal MP fibers correlated positively with fecal Bifidobacterium and negatively with Lachnospiraceae. Patients with MASH (n = 11) were re-assessed 12-months post-bariatric surgery (BSx) and we found that those with persistent disease (n = 4) had higher fecal MP fragments than those with normalized liver histology (n = 7). At 12-month post-BSx, MP fragments positively correlated with helper T cells and total MPs positively correlated with natural killer T cells and B cells. Our study is the first to look at 1) the role of MPs in MASH and its association with IM, immune cells and hepatic gene expression and 2) look at the role of MPs longitudinally in MASH persistence following BSx. Future research should further explore this relationship.
Collapse
Affiliation(s)
| | - Yasaman Ghorbani
- Toronto General Hospital, University Health Network, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada
| | | | - Lina Chen
- Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Kristina M Chomiak
- Thomas H. Gosnell School of Life Sciences, Rochester Institute of Technology, Rochester, NY, United States of America
| | - Anna Christina Tyler
- Thomas H. Gosnell School of Life Sciences, Rochester Institute of Technology, Rochester, NY, United States of America
| | - Nathan C Eddingsaas
- School of Chemistry and Materials Science, Rochester Institute of Technology, Rochester, NY, United States of America
| | - Sandra E Fischer
- Toronto General Hospital, University Health Network, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Timothy D Jackson
- Division of Surgery, University of Toronto, Toronto, Canada; Division of General Surgery, Toronto Western Hospital, University Health Network, Toronto, Canada
| | - Allan Okrainec
- Division of Surgery, University of Toronto, Toronto, Canada; Division of General Surgery, Toronto Western Hospital, University Health Network, Toronto, Canada
| | - Johane P Allard
- Toronto General Hospital, University Health Network, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada; Department of Medicine, Division of Gastroenterology, University of Toronto, Toronto, Canada.
| |
Collapse
|
13
|
De Ponti FF, Liu Z, Scott CL. Understanding the complex macrophage landscape in MASLD. JHEP Rep 2024; 6:101196. [PMID: 39524202 PMCID: PMC11550213 DOI: 10.1016/j.jhepr.2024.101196] [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: 07/01/2024] [Revised: 08/07/2024] [Accepted: 08/14/2024] [Indexed: 11/16/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) represents a spectrum of disease states ranging from simple steatosis to metabolic dysfunction-associated steatohepatitis (MASH), which can eventually lead to the development of cirrhosis and hepatocellular carcinoma. Macrophages have long been implicated in driving the progression from steatosis to end-stage disease, yet we still know relatively little about the precise involvement of these cells in MASLD progression and/or regression. Rather, there are a considerable number of conflicting reports regarding the precise roles of these cells. This confusion stems from the fact that, until recently, macrophages in the liver were considered a homogenous population. However, thanks to recent technological advances including multi-parameter flow cytometry, single-cell RNA sequencing and spatial proteogenomics, we now know that this is not the case. Rather hepatic macrophages, even in the healthy liver, are heterogenous, existing in multiple subsets with distinct transcriptional profiles and hence likely functions. This heterogeneity is even more prominent in MASLD, where the macrophage pool consists of multiple different subsets of resident and recruited cells. To probe the unique functions of these cells and determine if targeting macrophages may be a viable therapeutic strategy in MASLD, we first need to unravel this complexity and decipher which populations and/or activation states are present and what functions each of these may play in driving MASLD progression. In this review, we summarise recent advances in the field, highlighting what is currently known about the hepatic macrophage landscape in MASLD and the questions that remain to be tackled.
Collapse
Affiliation(s)
- Federico F. De Ponti
- Laboratory of Myeloid Cell Biology in Tissue Damage and Inflammation, VIB-UGent Center for Inflammation Research, Technologiepark-Zwijnaarde 71, Ghent 9052, Belgium
- Department of Biomedical Molecular Biology, Faculty of Science, Ghent University, Belgium
| | - Zhuangzhuang Liu
- Laboratory of Myeloid Cell Biology in Tissue Damage and Inflammation, VIB-UGent Center for Inflammation Research, Technologiepark-Zwijnaarde 71, Ghent 9052, Belgium
- Department of Biomedical Molecular Biology, Faculty of Science, Ghent University, Belgium
| | - Charlotte L. Scott
- Laboratory of Myeloid Cell Biology in Tissue Damage and Inflammation, VIB-UGent Center for Inflammation Research, Technologiepark-Zwijnaarde 71, Ghent 9052, Belgium
- Department of Biomedical Molecular Biology, Faculty of Science, Ghent University, Belgium
- Department of Chemical Sciences, Bernal Institute, University of Limerick, Castletroy, Co. Limerick, Ireland
| |
Collapse
|
14
|
Qian JK, Ma Y, Huang X, Li XR, Xu YF, Liu ZY, Gu Y, Shen K, Tian LJ, Wang YT, Cheng NN, Yang BS, Huang KY, Chai Y, Liu GQ, Cui NQ, Deng SY, Jiang N, Xu DR, Yu B. The CD163/TWEAK/Fn14 axis: A potential therapeutic target for alleviating inflammatory bone loss. J Orthop Translat 2024; 49:82-95. [PMID: 39430128 PMCID: PMC11488420 DOI: 10.1016/j.jot.2024.09.002] [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: 03/17/2024] [Revised: 08/21/2024] [Accepted: 09/01/2024] [Indexed: 10/22/2024] Open
Abstract
Objective Osteoclast (OC) over-activation is an important cause of bone loss that is strongly correlated with inflammation. Although the CD163/TWEAK/Fn14 axis has been implicated in several inflammatory pathologies, its contributions to inflammatory bone loss remain poorly understood. This study aimed to evaluate the interaction of the CD163/TWEAK/Fn14 axis with OC in inflammatory bone loss. Methods To assess the role of CD163 in bone homeostasis, we characterized the bone phenotypes of CD163-deficient mice and their wild-type littermates. CD163 and TWEAK levels were evaluated in the bone marrow of mice with LPS-induced bone loss and individuals with rheumatoid arthritis (RA). Bone mass changes were assessed using uCT and histology following supplementation with recombinant mouse CD163 protein (rCD163) or blockade of TWEAK/Fn14 signaling in CD163-deficient mice and mice with LPS-induced bone loss. The impact of CD163/TWEAK on OC differentiation and bone resorption capacity was analyzed in vitro. Results CD163 deficiency caused decreased bone mass and increased OC abundance. Lower CD163 expression and higher TWEAK expression were observed in the bone marrow of mice with LPS-induced bone loss and individuals with RA. TWEAK, mainly derived from CD68+ macrophages, was responsible for bone loss, and supplementing rCD163 or blocking TWEAK/Fn14 signaling contributed to rescue bone loss. TWEAK/Fn14 synergistically promoted RANKL-dependent OC differentiation and bone resorption capability through downstream mitogen-activated protein kinases (MAPK) signaling, while the pro-osteoclastic effect of TWEAK was suppressed by CD163. Conclusion Our findings suggest that the CD163/TWEAK/Fn14 axis is a potential therapeutic target for inflammatory bone loss by regulating osteoclastogenesis.
Collapse
Affiliation(s)
- Ji-kun Qian
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, The Seventh Affiliated Hospital, Southern Medical University, Foshan, 528200, China
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yuan Ma
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xuan Huang
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xiao-ran Li
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ya-fei Xu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, The Seventh Affiliated Hospital, Southern Medical University, Foshan, 528200, China
| | - Zi-ying Liu
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yuan Gu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ke Shen
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Liang-jie Tian
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yu-tian Wang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ning-ning Cheng
- Department of Obstetrics, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Bing-sheng Yang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Kui-yuan Huang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330000, China
| | - Yu Chai
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Guan-qiao Liu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Nai-qian Cui
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Song-yun Deng
- Department of Orthopaedics and Traumatology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Nan Jiang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Dao-rong Xu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Bin Yu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
15
|
Su T, He Y, Wang M, Zhou H, Huang Y, Ye M, Guo Q, Xiao Y, Cai G, Zhao M, Wang J, Luo X. Macrophage-Hepatocyte Circuits Mediated by Grancalcin Aggravate the Progression of Metabolic Dysfunction Associated Steatohepatitis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406500. [PMID: 39279458 PMCID: PMC11558151 DOI: 10.1002/advs.202406500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/16/2024] [Indexed: 09/18/2024]
Abstract
The dynamic interplay between parenchymal hepatocytes and non-parenchymal cells (NPCs), such as macrophages, is an important mechanism for liver metabolic homeostasis. Although numerous endeavors have been made to identify the mediators of metabolic dysfunction associated steatohepatitis (MASH), the molecular underpinnings of MASH progression remain poorly understood, and therapies to arrest MASH progression remain elusive. Herein, it is revealed that the expression of grancalcin (GCA) is upregulated in the macrophages of patients and rodents with MASH and correlates with MASH progression. Notably, the administration of recombinant GCA aggravates the development of MASH, whereas, Gca deletion in myeloid cells blunts liver steatosis and inflammation in multiple MASH murine models. Mechanistically, GCA activates macrophages via TLR9-NF-κB signaling, and the activated macrophages promote hepatocyte lipid accumulation and apoptosis via secretion of Interleukin-6(IL-6), Tumor Necrosis Factor α (TNFα), and Interleukin-1β(IL-1β), thereby leading to hepatic steatosis and inflammation. Finally, the therapeutic administration of antibody blocking GCA effectively halts the progression of MASH. Collectively, these findings implicate GCA as a crucial mediator of MASH and clarify a new metabolic signaling axis between the hepatocytes and macrophages, implying that GCA can emerge as a particularly interesting putative therapeutic target for reversing MASH progression.
Collapse
Affiliation(s)
- Tian Su
- Department of EndocrinologyEndocrinology Research CenterXiangya Hospital of Central South UniversityChangshaHunan410008China
| | - Yue He
- Department of EndocrinologyEndocrinology Research CenterXiangya Hospital of Central South UniversityChangshaHunan410008China
| | - Min Wang
- Department of EndocrinologyEndocrinology Research CenterXiangya Hospital of Central South UniversityChangshaHunan410008China
| | - Haiyan Zhou
- Department of EndocrinologyEndocrinology Research CenterXiangya Hospital of Central South UniversityChangshaHunan410008China
| | - Yan Huang
- Department of EndocrinologyEndocrinology Research CenterXiangya Hospital of Central South UniversityChangshaHunan410008China
| | - Mingsheng Ye
- Department of EndocrinologyEndocrinology Research CenterXiangya Hospital of Central South UniversityChangshaHunan410008China
| | - Qi Guo
- Department of EndocrinologyEndocrinology Research CenterXiangya Hospital of Central South UniversityChangshaHunan410008China
| | - Ye Xiao
- Department of EndocrinologyEndocrinology Research CenterXiangya Hospital of Central South UniversityChangshaHunan410008China
| | - Guangping Cai
- Department of EndocrinologyEndocrinology Research CenterXiangya Hospital of Central South UniversityChangshaHunan410008China
| | - Mingyang Zhao
- Department of EndocrinologyEndocrinology Research CenterXiangya Hospital of Central South UniversityChangshaHunan410008China
| | - Jianping Wang
- Department of EndocrinologyThe Second Affiliated Hospital of University of South ChinaHengyangHunan421000China
| | - Xianghang Luo
- Department of EndocrinologyEndocrinology Research CenterXiangya Hospital of Central South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalChangshaHunan410008China
| |
Collapse
|
16
|
Khanna K, Eul E, Yan H, Faccio R. Delayed macrophage targeting by clodronate liposomes worsens the progression of cytokine storm syndrome. Front Immunol 2024; 15:1477449. [PMID: 39530102 PMCID: PMC11550973 DOI: 10.3389/fimmu.2024.1477449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
Excessive macrophage activation and production of pro-inflammatory cytokines are hallmarks of the Cytokine Storm Syndrome (CSS), a lethal condition triggered by sepsis, autoimmune disorders, and cancer immunotherapies. While depletion of macrophages at disease onset protects from lethality in an infection-induced CSS murine model, patients are rarely diagnosed early, hence the need to characterize macrophage populations during CSS progression and assess the therapeutic implications of macrophage targeting after disease onset. In this study, we identified MHCII+F4/80+Tim4- macrophages as the primary contributors to the pro-inflammatory environment in CSS, while CD206+F4/80+Tim4+ macrophages, with an anti-inflammatory profile, become outnumbered. Additionally, we observed an expansion of Tim4- macrophages coinciding with increased hematopoietic stem progenitor cells and reduction of committed myeloid progenitors in bone marrow and spleen. Critically, macrophage targeting with clodronate liposomes at disease onset prolonged survival, while their targeting in mice with established CSS exacerbated disease severity, leading to a more dramatic loss of Tim4+ macrophages and an imbalance in pro- versus anti-inflammatory Tim4- macrophage ratio. Our findings highlight the significance of timing in macrophage-targeted interventions for effective management of CSS and suggest potential therapeutic strategies for diseases characterized by uncontrolled inflammation, such as sepsis.
Collapse
Affiliation(s)
- Kunjan Khanna
- Department of Orthopedics, Washington University in St Louis, St Louis, MO, United States
| | - Emily Eul
- Department of Orthopedics, Washington University in St Louis, St Louis, MO, United States
| | - Hui Yan
- Department of Orthopedics, Washington University in St Louis, St Louis, MO, United States
| | - Roberta Faccio
- Department of Orthopedics, Washington University in St Louis, St Louis, MO, United States
- Shriners Hospital for Children, St Louis, MO, United States
| |
Collapse
|
17
|
Tsourmas KI, Butler CA, Kwang NE, Sloane ZR, Dykman KJG, Maloof GO, Prekopa CA, Krattli RP, El-Khatib SM, Swarup V, Acharya MM, Hohsfield LA, Green KN. Myeloid-derived β-hexosaminidase is essential for neuronal health and lysosome function: implications for Sandhoff disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.21.619538. [PMID: 39484433 PMCID: PMC11526954 DOI: 10.1101/2024.10.21.619538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Lysosomal storage disorders (LSDs) are a large disease class involving lysosomal dysfunction, often resulting in neurodegeneration. Sandhoff disease (SD) is an LSD caused by a deficiency in the β subunit of the β-hexosaminidase enzyme (Hexb). Although Hexb expression in the brain is specific to microglia, SD primarily affects neurons. To understand how a microglial gene is involved in maintaining neuronal homeostasis, we demonstrated that β-hexosaminidase is secreted by microglia and integrated into the neuronal lysosomal compartment. To assess therapeutic relevance, we treated SD mice with bone marrow transplant and colony stimulating factor 1 receptor inhibition, which broadly replaced Hexb -/- microglia with Hexb-sufficient cells. This intervention reversed apoptotic gene signatures, improved behavior, restored enzymatic activity and Hexb expression, ameliorated substrate accumulation, and normalized neuronal lysosomal phenotypes. These results underscore the critical role of myeloid-derived β-hexosaminidase in neuronal lysosomal function and establish microglial replacement as a potential LSD therapy.
Collapse
Affiliation(s)
- Kate I. Tsourmas
- Department of Neurobiology and Behavior; University of California; Irvine, CA 92697; USA
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine, CA 92697; USA
| | - Claire A. Butler
- Department of Neurobiology and Behavior; University of California; Irvine, CA 92697; USA
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine, CA 92697; USA
| | - Nellie E. Kwang
- Department of Neurobiology and Behavior; University of California; Irvine, CA 92697; USA
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine, CA 92697; USA
| | - Zachary R. Sloane
- Department of Neurobiology and Behavior; University of California; Irvine, CA 92697; USA
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine, CA 92697; USA
| | - Koby J. G. Dykman
- Department of Neurobiology and Behavior; University of California; Irvine, CA 92697; USA
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine, CA 92697; USA
| | - Ghassan O. Maloof
- Department of Neurobiology and Behavior; University of California; Irvine, CA 92697; USA
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine, CA 92697; USA
| | - Christiana A. Prekopa
- Department of Neurobiology and Behavior; University of California; Irvine, CA 92697; USA
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine, CA 92697; USA
| | - Robert P. Krattli
- Department of Anatomy and Neurobiology; University of California; Irvine, CA 92697; USA
| | - Sanad M. El-Khatib
- Department of Anatomy and Neurobiology; University of California; Irvine, CA 92697; USA
| | - Vivek Swarup
- Department of Neurobiology and Behavior; University of California; Irvine, CA 92697; USA
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine, CA 92697; USA
| | - Munjal M. Acharya
- Department of Anatomy and Neurobiology; University of California; Irvine, CA 92697; USA
- Department of Radiation Oncology; University of California; Irvine, CA 92697; USA
| | - Lindsay A. Hohsfield
- Department of Neurobiology and Behavior; University of California; Irvine, CA 92697; USA
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine, CA 92697; USA
| | - Kim N. Green
- Department of Neurobiology and Behavior; University of California; Irvine, CA 92697; USA
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine, CA 92697; USA
| |
Collapse
|
18
|
Han D, Ma Q, Ballar P, Zhang C, Dai M, Luo X, Gu J, Wei C, Guo P, Zeng L, Hu M, Jiang C, Liang Y, Wang Y, Hou C, Wang X, Feng L, Shen Y, Shen Y, Hu X, Liu J. Reprogramming tumor-associated macrophages and inhibiting tumor neovascularization by targeting MANF-HSF1-HSP70-1 pathway: An effective treatment for hepatocellular carcinoma. Acta Pharm Sin B 2024; 14:4396-4412. [PMID: 39525584 PMCID: PMC11544390 DOI: 10.1016/j.apsb.2024.05.001] [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: 01/26/2024] [Revised: 04/09/2024] [Accepted: 04/26/2024] [Indexed: 11/16/2024] Open
Abstract
In advanced hepatocellular carcinoma (HCC) tissues, M2-like tumor-associated macrophages (TAMs) are in the majority and promotes HCC progression. Contrary to the pro-tumor effect of M2-like TAMs, M1-like TAMs account for a small proportion and have anti-tumor effects. Since TAMs can switch from one type to another, reprogramming TAMs may be an important treatment for HCC therapy. However, the mechanisms of phenotypic switch and reprogramming TAMs are still obscure. In this study, we analyzed differential genes in normal macrophages and TAMs, and found that loss of MANF in TAMs accompanied by high levels of downstream genes negatively regulated by MANF. MANF reprogrammed TAMs into M1 phenotype. Meanwhile, loss of MANF promoted HCC progression in HCC patients and mice HCC model, especially tumor neovascularization. Additionally, macrophages with MANF supplement suppressed HCC progression in mice, suggesting MANF supplement in macrophage was an effective treatment for HCC. Mechanistically, MANF enhanced the HSF1-HSP70-1 interaction, restricted HSF1 in the cytoplasm of macrophages, and decreased both mRNA and protein levels of HSP70-1, which in turn led to reprogramming TAMs, and suppressing neovascularization of HCC. Our study contributes to the exploration the mechanism of TAMs reprogramming, which may provide insights for future therapeutic exploitation of HCC neovascularization.
Collapse
Affiliation(s)
- Dan Han
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei 230032, China
| | - Qiannan Ma
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei 230032, China
| | - Petek Ballar
- Department of Biochemistry, Faculty of Pharmacy, Ege University, Izmir 35130, Turkey
| | - Chunyang Zhang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei 230032, China
| | - Min Dai
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei 230032, China
| | - Xiaoyuan Luo
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei 230032, China
| | - Jiong Gu
- Department of General Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei 230061, China
| | - Chuansheng Wei
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei 230032, China
| | - Panhui Guo
- Department of Gastroenterology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230061, China
| | - Lulu Zeng
- Department of Gastroenterology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230061, China
| | - Min Hu
- Department of Gastroenterology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230061, China
| | - Can Jiang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei 230032, China
| | - Yanyan Liang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei 230032, China
| | - Yanyan Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei 230032, China
| | - Chao Hou
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Xian Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Lijie Feng
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei 230032, China
| | - Yujun Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei 230032, China
| | - Yuxian Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei 230032, China
| | - Xiangpeng Hu
- Department of Gastroenterology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230061, China
| | - Jun Liu
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
19
|
Fima R, Dussaud S, Benbida C, Blanchet M, Lanthiez F, Poupel L, Brambilla C, Gélineau A, Dessena M, Blanc M, Lerévérend C, Moreau M, Boissonnas A, Gautier EL, Huby T. Loss of embryonically-derived Kupffer cells during hypercholesterolemia accelerates atherosclerosis development. Nat Commun 2024; 15:8341. [PMID: 39333539 PMCID: PMC11436809 DOI: 10.1038/s41467-024-52735-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 09/18/2024] [Indexed: 09/29/2024] Open
Abstract
Hypercholesterolemia is a major risk factor for atherosclerosis and associated cardiovascular diseases. The liver plays a key role in the regulation of plasma cholesterol levels and hosts a large population of tissue-resident macrophages known as Kupffer cells (KCs). KCs are located in the hepatic sinusoids where they ensure key functions including blood immune surveillance. However, how KCs homeostasis is affected by the build-up of cholesterol-rich lipoproteins that occurs in the circulation during hypercholesterolemia remains unknown. Here, we show that embryo-derived KCs (EmKCs) accumulate large amounts of lipoprotein-derived cholesterol, in part through the scavenger receptor CD36, and massively expand early after the induction of hypercholesterolemia. After this rapid adaptive response, EmKCs exhibit mitochondrial oxidative stress and their numbers gradually diminish while monocyte-derived KCs (MoKCs) with reduced cholesterol-loading capacities seed the KC pool. Decreased proportion of EmKCs in the KC pool enhances liver cholesterol content and exacerbates hypercholesterolemia, leading to accelerated atherosclerotic plaque development. Together, our data reveal that KC homeostasis is perturbed during hypercholesterolemia, which in turn alters the control of plasma cholesterol levels and increases atherosclerosis.
Collapse
Affiliation(s)
- Rebecca Fima
- Sorbonne Université, INSERM, UMRS 1166, 75013, Paris, France
| | | | - Cheïma Benbida
- Sorbonne Université, INSERM, UMRS 1166, 75013, Paris, France
| | | | - François Lanthiez
- INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, Sorbonne Université, 75013, Paris, France
| | - Lucie Poupel
- Sorbonne Université, INSERM, UMRS 1166, 75013, Paris, France
| | | | | | - Mattia Dessena
- Sorbonne Université, INSERM, UMRS 1166, 75013, Paris, France
- Department of Medicine and Surgery, University of Parma, 43125, Parma, Italy
| | - Marina Blanc
- Sorbonne Université, INSERM, UMRS 1166, 75013, Paris, France
| | | | - Martine Moreau
- Sorbonne Université, INSERM, UMRS 1166, 75013, Paris, France
| | - Alexandre Boissonnas
- INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, Sorbonne Université, 75013, Paris, France
| | | | - Thierry Huby
- Sorbonne Université, INSERM, UMRS 1166, 75013, Paris, France.
| |
Collapse
|
20
|
Ruiz-Blázquez P, Fernández-Fernández M, Pistorio V, Martinez-Sanchez C, Costanzo M, Iruzubieta P, Zhuravleva E, Cacho-Pujol J, Ariño S, Del Castillo-Cruz A, Núñez S, Andersen JB, Ruoppolo M, Crespo J, García-Ruiz C, Pavone LM, Reinheckel T, Sancho-Bru P, Coll M, Fernández-Checa JC, Moles A. Cathepsin D is essential for the degradomic shift of macrophages required to resolve liver fibrosis. Mol Metab 2024; 87:101989. [PMID: 39019115 PMCID: PMC11327474 DOI: 10.1016/j.molmet.2024.101989] [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: 05/14/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Fibrosis contributes to 45% of deaths in industrialized nations and is characterized by an abnormal accumulation of extracellular matrix (ECM). There are no specific anti-fibrotic treatments for liver fibrosis, and previous unsuccessful attempts at drug development have focused on preventing ECM deposition. Because liver fibrosis is largely acknowledged to be reversible, regulating fibrosis resolution could offer novel therapeutical options. However, little is known about the mechanisms controlling ECM remodeling during resolution. Changes in proteolytic activity are essential for ECM homeostasis and macrophages are an important source of proteases. Herein, in this study we evaluate the role of macrophage-derived cathepsin D (CtsD) during liver fibrosis. METHODS CtsD expression and associated pathways were characterized in single-cell RNA sequencing and transcriptomic datasets in human cirrhosis. Liver fibrosis progression, reversion and functional characterization were assessed in novel myeloid-CtsD and hepatocyte-CtsD knock-out mice. RESULTS Analysis of single-cell RNA sequencing datasets demonstrated CtsD was expressed in macrophages and hepatocytes in human cirrhosis. Liver fibrosis progression, reversion and functional characterization were assessed in novel myeloid-CtsD (CtsDΔMyel) and hepatocyte-CtsD knock-out mice. CtsD deletion in macrophages, but not in hepatocytes, resulted in enhanced liver fibrosis. Both inflammatory and matrisome proteomic signatures were enriched in fibrotic CtsDΔMyel livers. Besides, CtsDΔMyel liver macrophages displayed functional, phenotypical and secretomic changes, which resulted in a degradomic phenotypical shift, responsible for the defective proteolytic processing of collagen I in vitro and impaired collagen remodeling during fibrosis resolution in vivo. Finally, CtsD-expressing mononuclear phagocytes of cirrhotic human livers were enriched in lysosomal and ECM degradative signaling pathways. CONCLUSIONS Our work describes for the first-time CtsD-driven lysosomal activity as a central hub for restorative macrophage function during fibrosis resolution and opens new avenues to explore their degradome landscape to inform drug development.
Collapse
Affiliation(s)
- Paloma Ruiz-Blázquez
- Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain; CiberEHD, Spain; University of Barcelona, Barcelona, Spain; IDIBAPS, Barcelona, Spain
| | - María Fernández-Fernández
- Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain; CiberEHD, Spain; University of Barcelona, Barcelona, Spain; IDIBAPS, Barcelona, Spain
| | - Valeria Pistorio
- Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain; Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy; Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine (CRSA), Paris, France
| | | | - Michele Costanzo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy; CEINGE-Biotecnologie Avanzate Franco Salvatore s.c.ar.l., Naples, Italy
| | - Paula Iruzubieta
- Department of Gastroenterology and Hepatology, Marqués de Valdecilla University Hospital, Research Institute Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - Ekaterina Zhuravleva
- Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; LEO Foundation Skin Immunology Research Center (SIC), Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Júlia Cacho-Pujol
- Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain; University of Barcelona, Barcelona, Spain; IDIBAPS, Barcelona, Spain
| | - Silvia Ariño
- CiberEHD, Spain; University of Barcelona, Barcelona, Spain; IDIBAPS, Barcelona, Spain
| | | | | | - Jesper B Andersen
- Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Margherita Ruoppolo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy; CEINGE-Biotecnologie Avanzate Franco Salvatore s.c.ar.l., Naples, Italy
| | - Javier Crespo
- Department of Gastroenterology and Hepatology, Marqués de Valdecilla University Hospital, Research Institute Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - Carmen García-Ruiz
- Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain; CiberEHD, Spain; IDIBAPS, Barcelona, Spain; USC Research Center for ALPD, Los Angeles, United States; Associated Unit IIBB-IMIM, Barcelona, Spain
| | - Luigi Michele Pavone
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Thomas Reinheckel
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Albert-Ludwigs-University, Freiburg, Germany; German Cancer Consortium (DKTK), DKFZ Partner Site Freiburg, Germany; Center for Biological Signaling Studies BIOSS, University of Freiburg, Germany
| | - Pau Sancho-Bru
- CiberEHD, Spain; University of Barcelona, Barcelona, Spain; IDIBAPS, Barcelona, Spain
| | - Mar Coll
- CiberEHD, Spain; University of Barcelona, Barcelona, Spain; IDIBAPS, Barcelona, Spain; Medicine Department, Faculty of Medicine, University of Barcelona, Spain
| | - José C Fernández-Checa
- Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain; CiberEHD, Spain; IDIBAPS, Barcelona, Spain; USC Research Center for ALPD, Los Angeles, United States; Associated Unit IIBB-IMIM, Barcelona, Spain
| | - Anna Moles
- Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain; CiberEHD, Spain; IDIBAPS, Barcelona, Spain; Associated Unit IIBB-IMIM, Barcelona, Spain.
| |
Collapse
|
21
|
Doyle EH, Vaughan HJ, Mariani SA. From drosophila to humans: a journey through macrophage development. Exp Hematol 2024; 136:104272. [PMID: 38972565 DOI: 10.1016/j.exphem.2024.104272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/28/2024] [Accepted: 06/30/2024] [Indexed: 07/09/2024]
Abstract
Macrophages are fascinating immune cells involved in a variety of processes in both health and disease. Although they were first discovered and characterized by their functions as professional phagocytes and antigen-presenting cells, it is now clear that macrophages have multiple roles within embryonic development, tissue homeostasis, regulation of inflammation, and host response to pathogens and tissue insults. Interestingly, macrophages, or macrophage-like cells, exist in a variety of organisms, from echinoderms to humans, and are present also in species that lack an adaptive immune system or hematopoietic stem cells (HSCs). In mammals, macrophages can be generated from bone marrow precursors through a monocyte intermediate, but it is now known that they are also generated during earlier hematopoietic waves in the embryo. Seeding a variety of tissues at different times, macrophages contribute to embryonic organogenesis and tissue homeostasis. Interestingly, in species where embryonic macrophages are generated before HSC specification, they seem to be an important component of the HSC generative microenvironment. There are many excellent reviews reporting the current knowledge on the ontogeny and functions of macrophages in adult tissues. Here, we aim to summarize the current knowledge on the development and functions of embryonic macrophages across the most used animal models, with a special focus on developmental hematopoiesis.
Collapse
Affiliation(s)
- Eva H Doyle
- Centre for Inflammation Research, Institute of Regeneration and Repair, The University of Edinburgh, Edinburgh, United Kingdom
| | - Hollie J Vaughan
- Centre for Inflammation Research, Institute of Regeneration and Repair, The University of Edinburgh, Edinburgh, United Kingdom
| | - Samanta A Mariani
- Centre for Inflammation Research, Institute of Regeneration and Repair, The University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
22
|
Pessenda G, Ferreira TR, Paun A, Kabat J, Amaral EP, Kamenyeva O, Gazzinelli-Guimaraes PH, Perera SR, Ganesan S, Hun Lee S, Sacks DL. Kupffer cell and recruited macrophage heterogeneity orchestrate granuloma maturation and hepatic immunity in visceral leishmaniasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.09.602717. [PMID: 39372777 PMCID: PMC11451627 DOI: 10.1101/2024.07.09.602717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
In murine models of visceral leishmaniasis (VL), parasitization of resident Kupffer cells (resKCs) is responsible for early growth of Leishmania infantum in the liver, which leads to granuloma formation and eventual parasite control. We employed the chronic VL model, and revealed an open niche established by KCs death and their migration outside of the sinusoids, resulting in their gradual replacement by monocyte-derived KCs (moKCs). While early granulomas were composed of resKCs, late granulomas were found outside of the sinusoids and contained resKC-derived macrophages, and monocyte-derived macrophages (momacs). ResKCs and moKCs within the sinusoids were identified as homeostatic/regulatory cells, while resKC-derived macrophages and momacs within late granulomas were pro-inflammatory. Despite the infection being largely confined to the resKC-derived macrophages, in the absence of monocyte recruitment, parasite control was strongly compromised. Macrophage heterogeneity, involving migration and reprogramming of resKCs, along with recruitment of monocyte-derived cells, is a hallmark of granuloma maturation and hepatic immunity in VL.
Collapse
Affiliation(s)
- Gabriela Pessenda
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tiago R. Ferreira
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Andrea Paun
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Juraj Kabat
- Biological Imaging Section, Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Eduardo P. Amaral
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Olena Kamenyeva
- Biological Imaging Section, Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Pedro Henrique Gazzinelli-Guimaraes
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shehan R. Perera
- Department of Electrical and Computer Engineering, The Ohio State University, Columbus, OH 43201, USA
| | - Sundar Ganesan
- Biological Imaging Section, Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sang Hun Lee
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - David L. Sacks
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
23
|
Wu Y, Riehle A, Pollmeier B, Kadow S, Schumacher F, Drab M, Kleuser B, Gulbins E, Grassmé H. Caveolin-1 affects early mycobacterial infection and apoptosis in macrophages and mice. Tuberculosis (Edinb) 2024; 147:102493. [PMID: 38547568 DOI: 10.1016/j.tube.2024.102493] [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: 10/16/2023] [Revised: 02/09/2024] [Accepted: 02/11/2024] [Indexed: 06/14/2024]
Abstract
Tuberculosis, caused by Mycobacterium tuberculosis, remains one of the deadliest infections in humans. Because Mycobacterium bovis Bacillus Calmette-Guérin (BCG) share genetic similarities with Mycobacterium tuberculosis, it is often used as a model to elucidate the molecular mechanisms of more severe tuberculosis infection. Caveolin-1 has been implied in many physiological processes and diseases, but it's role in mycobacterial infections has barely been studied. We isolated macrophages from Wildtype or Caveolin-1 deficient mice and analyzed hallmarks of infection, such as internalization, induction of autophagy and apoptosis. For in vivo assays we intravenously injected mice with BCG and investigated tissues for bacterial load with colony-forming unit assays, bioactive lipids with mass spectrometry and changes of protein expressions by Western blotting. Our results revealed that Caveolin-1 was important for early killing of BCG infection in vivo and in vitro, controlled acid sphingomyelinase (Asm)-dependent ceramide formation, apoptosis and inflammatory cytokines upon infection with BCG. In accordance, Caveolin-1 deficient mice and macrophages showed higher bacterial burdens in the livers. The findings indicate that Caveolin-1 plays a role in infection of mice and murine macrophages with BCG, by controlling cellular apoptosis and inflammatory host response. These clues might be useful in the fight against tuberculosis.
Collapse
Affiliation(s)
- Yuqing Wu
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Andrea Riehle
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Barbara Pollmeier
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Stephanie Kadow
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | | | - Marek Drab
- Unit of Nanostructural Biointeractions, Department of Immunology of Infectious Diseases, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 12 Weigla Street, 53-114, Wroclaw, Poland
| | - Burkhard Kleuser
- Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Erich Gulbins
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Heike Grassmé
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany.
| |
Collapse
|
24
|
Tighe RM, Birukova A, Malakhau Y, Kobayashi Y, Vose AT, Chandramohan V, Cyphert-Daly JM, Cumming RI, Fradin Kirshner H, Tata PR, Ingram JL, Gunn MD, Que LG, Yu YRA. Altered ontogeny and transcriptomic signatures of tissue-resident pulmonary interstitial macrophages ameliorate allergic airway hyperresponsiveness. Front Immunol 2024; 15:1371764. [PMID: 38983858 PMCID: PMC11231371 DOI: 10.3389/fimmu.2024.1371764] [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: 01/16/2024] [Accepted: 06/05/2024] [Indexed: 07/11/2024] Open
Abstract
Introduction Environmental exposures and experimental manipulations can alter the ontogenetic composition of tissue-resident macrophages. However, the impact of these alterations on subsequent immune responses, particularly in allergic airway diseases, remains poorly understood. This study aims to elucidate the significance of modified macrophage ontogeny resulting from environmental exposures on allergic airway responses to house dust mite (HDM) allergen. Methods We utilized embryonic lineage labeling to delineate the ontogenetic profile of tissue-resident macrophages at baseline and following the resolution of repeated lipopolysaccharide (LPS)-induced lung injury. We investigated differences in house dust mite (HDM)-induced allergy to assess the influence of macrophage ontogeny on allergic airway responses. Additionally, we employed single-cell RNA sequencing (scRNAseq) and immunofluorescent staining to characterize the pulmonary macrophage composition, associated pathways, and tissue localization. Results Our findings demonstrate that the ontogeny of homeostatic alveolar and interstitial macrophages is altered after the resolution from repeated LPS-induced lung injury, leading to the replacement of embryonic-derived by bone marrow-derived macrophages. This shift in macrophage ontogeny is associated with reduced HDM-induced allergic airway responses. Through scRNAseq and immunofluorescent staining, we identified a distinct subset of resident-derived interstitial macrophages expressing genes associated with allergic airway diseases, localized adjacent to terminal bronchi, and diminished by prior LPS exposure. Discussion These results suggest a pivotal role for pulmonary macrophage ontogeny in modulating allergic airway responses. Moreover, our findings highlight the implications of prior environmental exposures in shaping future immune responses and influencing the development of allergies. By elucidating the mechanisms underlying these phenomena, this study provides valuable insights into potential therapeutic targets for allergic airway diseases and avenues for further research into immune modulation and allergic disease prevention.
Collapse
Affiliation(s)
- Robert M. Tighe
- Department of Medicine, Duke University, Durham, NC, United States
| | | | - Yuryi Malakhau
- Department of Medicine, Duke University, Durham, NC, United States
| | - Yoshihiko Kobayashi
- Department of Cell Biology, Duke University, Durham, NC, United States
- Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Aaron T. Vose
- Department of Medicine, Duke University, Durham, NC, United States
| | | | | | - R. Ian Cumming
- Department of Medicine, Duke University, Durham, NC, United States
| | | | | | | | - Michael D. Gunn
- Department of Medicine, Duke University, Durham, NC, United States
| | - Loretta G. Que
- Department of Medicine, Duke University, Durham, NC, United States
| | - Yen-Rei A. Yu
- Department of Medicine, Duke University, Durham, NC, United States
- Department of Medicine, University of Colorado Anschutz School of Medicine, Aurora, CO, United States
| |
Collapse
|
25
|
Arlat A, Renoud ML, Nakhle J, Thomas M, Fontaine J, Arnaud E, Dray C, Authier H, Monsarrat P, Coste A, Casteilla L, Ousset M, Cousin B. Generation of functionally active resident macrophages from adipose tissue by 3D cultures. Front Immunol 2024; 15:1356397. [PMID: 38975341 PMCID: PMC11224291 DOI: 10.3389/fimmu.2024.1356397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 05/31/2024] [Indexed: 07/09/2024] Open
Abstract
Introduction Within adipose tissue (AT), different macrophage subsets have been described, which played pivotal and specific roles in upholding tissue homeostasis under both physiological and pathological conditions. Nonetheless, studying resident macrophages in-vitro poses challenges, as the isolation process and the culture for extended periods can alter their inherent properties. Methods Stroma-vascular cells isolated from murine subcutaneous AT were seeded on ultra-low adherent plates in the presence of macrophage colony-stimulating factor. After 4 days of culture, the cells spontaneously aggregate to form spheroids. A week later, macrophages begin to spread out of the spheroid and adhere to the culture plate. Results This innovative three-dimensional (3D) culture method enables the generation of functional mature macrophages that present distinct genic and phenotypic characteristics compared to bone marrow-derived macrophages. They also show specific metabolic activity and polarization in response to stimulation, but similar phagocytic capacity. Additionally, based on single-cell analysis, AT-macrophages generated in 3D culture mirror the phenotypic and functional traits of in-vivo AT resident macrophages. Discussion Our study describes a 3D in-vitro system for generating and culturing functional AT-resident macrophages, without the need for cell sorting. This system thus stands as a valuable resource for exploring the differentiation and function of AT-macrophages in vitro in diverse physiological and pathological contexts.
Collapse
Affiliation(s)
- Adèle Arlat
- RESTORE Research Center, Université de Toulouse, INSERM 1301, CNRS 5070, Etablissement Français du Sang (EFS), Ecole Nationale Vétérinaire de Touloue (ENVT), Toulouse, France
| | - Marie-Laure Renoud
- RESTORE Research Center, Université de Toulouse, INSERM 1301, CNRS 5070, Etablissement Français du Sang (EFS), Ecole Nationale Vétérinaire de Touloue (ENVT), Toulouse, France
| | - Jean Nakhle
- RESTORE Research Center, Université de Toulouse, INSERM 1301, CNRS 5070, Etablissement Français du Sang (EFS), Ecole Nationale Vétérinaire de Touloue (ENVT), Toulouse, France
| | - Miguel Thomas
- RESTORE Research Center, Université de Toulouse, INSERM 1301, CNRS 5070, Etablissement Français du Sang (EFS), Ecole Nationale Vétérinaire de Touloue (ENVT), Toulouse, France
| | - Jessica Fontaine
- RESTORE Research Center, Université de Toulouse, INSERM 1301, CNRS 5070, Etablissement Français du Sang (EFS), Ecole Nationale Vétérinaire de Touloue (ENVT), Toulouse, France
| | - Emmanuelle Arnaud
- RESTORE Research Center, Université de Toulouse, INSERM 1301, CNRS 5070, Etablissement Français du Sang (EFS), Ecole Nationale Vétérinaire de Touloue (ENVT), Toulouse, France
| | - Cédric Dray
- RESTORE Research Center, Université de Toulouse, INSERM 1301, CNRS 5070, Etablissement Français du Sang (EFS), Ecole Nationale Vétérinaire de Touloue (ENVT), Toulouse, France
| | - Hélène Authier
- RESTORE Research Center, Université de Toulouse, INSERM 1301, CNRS 5070, Etablissement Français du Sang (EFS), Ecole Nationale Vétérinaire de Touloue (ENVT), Toulouse, France
| | - Paul Monsarrat
- RESTORE Research Center, Université de Toulouse, INSERM 1301, CNRS 5070, Etablissement Français du Sang (EFS), Ecole Nationale Vétérinaire de Touloue (ENVT), Toulouse, France
- Dental Faculty and Hospital of Toulouse – Toulouse Institute of Oral Medicine and Science, CHU de Toulouse, Toulouse, France
- Artificial and Natural Intelligence Toulouse Institute (ANITI), Toulouse, France
| | - Agnès Coste
- RESTORE Research Center, Université de Toulouse, INSERM 1301, CNRS 5070, Etablissement Français du Sang (EFS), Ecole Nationale Vétérinaire de Touloue (ENVT), Toulouse, France
| | - Louis Casteilla
- RESTORE Research Center, Université de Toulouse, INSERM 1301, CNRS 5070, Etablissement Français du Sang (EFS), Ecole Nationale Vétérinaire de Touloue (ENVT), Toulouse, France
| | - Marielle Ousset
- RESTORE Research Center, Université de Toulouse, INSERM 1301, CNRS 5070, Etablissement Français du Sang (EFS), Ecole Nationale Vétérinaire de Touloue (ENVT), Toulouse, France
| | - Béatrice Cousin
- RESTORE Research Center, Université de Toulouse, INSERM 1301, CNRS 5070, Etablissement Français du Sang (EFS), Ecole Nationale Vétérinaire de Touloue (ENVT), Toulouse, France
| |
Collapse
|
26
|
Zelepukin IV, Shevchenko KG, Deyev SM. Rediscovery of mononuclear phagocyte system blockade for nanoparticle drug delivery. Nat Commun 2024; 15:4366. [PMID: 38777821 PMCID: PMC11111695 DOI: 10.1038/s41467-024-48838-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 05/14/2024] [Indexed: 05/25/2024] Open
Abstract
Rapid uptake of nanoparticles by mononuclear phagocyte system (MPS) significantly hampers their therapeutic efficacy. Temporal MPS blockade is one of the few ways to overcome this barrier - the approach rediscovered many times under different names but never extensively used in clinic. Using meta-analysis of the published data we prove the efficacy of this technique for enhancing particle circulation in blood and their delivery to tumours, describe a century of its evolution and potential combined mechanism behind it. Finally, we discuss future directions of the research focusing on the features essential for successful clinical translation of the method.
Collapse
Affiliation(s)
- Ivan V Zelepukin
- Department of Medicinal Chemistry, Uppsala University, 751 23, Uppsala, Sweden.
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997, Moscow, Russia.
| | | | - Sergey M Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997, Moscow, Russia
| |
Collapse
|
27
|
Miyamoto Y, Kikuta J, Matsui T, Hasegawa T, Fujii K, Okuzaki D, Liu YC, Yoshioka T, Seno S, Motooka D, Uchida Y, Yamashita E, Kobayashi S, Eguchi H, Morii E, Tryggvason K, Shichita T, Kayama H, Atarashi K, Kunisawa J, Honda K, Takeda K, Ishii M. Periportal macrophages protect against commensal-driven liver inflammation. Nature 2024; 629:901-909. [PMID: 38658756 DOI: 10.1038/s41586-024-07372-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 04/02/2024] [Indexed: 04/26/2024]
Abstract
The liver is the main gateway from the gut, and the unidirectional sinusoidal flow from portal to central veins constitutes heterogenous zones, including the periportal vein (PV) and the pericentral vein zones1-5. However, functional differences in the immune system in each zone remain poorly understood. Here intravital imaging revealed that inflammatory responses are suppressed in PV zones. Zone-specific single-cell transcriptomics detected a subset of immunosuppressive macrophages enriched in PV zones that express high levels of interleukin-10 and Marco, a scavenger receptor that sequesters pro-inflammatory pathogen-associated molecular patterns and damage-associated molecular patterns, and consequently suppress immune responses. Induction of Marco+ immunosuppressive macrophages depended on gut microbiota. In particular, a specific bacterial family, Odoribacteraceae, was identified to induce this macrophage subset through its postbiotic isoallolithocholic acid. Intestinal barrier leakage resulted in inflammation in PV zones, which was markedly augmented in Marco-deficient conditions. Chronic liver inflammatory diseases such as primary sclerosing cholangitis (PSC) and non-alcoholic steatohepatitis (NASH) showed decreased numbers of Marco+ macrophages. Functional ablation of Marco+ macrophages led to PSC-like inflammatory phenotypes related to colitis and exacerbated steatosis in NASH in animal experimental models. Collectively, commensal bacteria induce Marco+ immunosuppressive macrophages, which consequently limit excessive inflammation at the gateway of the liver. Failure of this self-limiting system promotes hepatic inflammatory disorders such as PSC and NASH.
Collapse
Affiliation(s)
- Yu Miyamoto
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Life-omics Research Division, Institute for Open and Transdisciplinary Research Initiative, Osaka University, Osaka, Japan
| | - Junichi Kikuta
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Life-omics Research Division, Institute for Open and Transdisciplinary Research Initiative, Osaka University, Osaka, Japan
- Laboratory of Bioimaging and Drug Discovery, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Takahiro Matsui
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- Department of Pathology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tetsuo Hasegawa
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
| | - Kentaro Fujii
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Life-omics Research Division, Institute for Open and Transdisciplinary Research Initiative, Osaka University, Osaka, Japan
| | - Daisuke Okuzaki
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Yu-Chen Liu
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Takuya Yoshioka
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Shigeto Seno
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, Osaka, Japan
| | - Daisuke Motooka
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Yutaka Uchida
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Life-omics Research Division, Institute for Open and Transdisciplinary Research Initiative, Osaka University, Osaka, Japan
- Laboratory of Bioimaging and Drug Discovery, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Erika Yamashita
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Life-omics Research Division, Institute for Open and Transdisciplinary Research Initiative, Osaka University, Osaka, Japan
| | - Shogo Kobayashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Eiichi Morii
- Department of Pathology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Karl Tryggvason
- Cardiovascular and Metabolic Disorders Program, Duke-NUS, Duke-NUS Medical School, Singapore, Singapore
| | - Takashi Shichita
- Laboratory for Neuroinflammation and Repair, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hisako Kayama
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Koji Atarashi
- Department of Microbiology and Immunology, School of Medicine, Keio University, Tokyo, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Kenya Honda
- Department of Microbiology and Immunology, School of Medicine, Keio University, Tokyo, Japan
| | - Kiyoshi Takeda
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Masaru Ishii
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan.
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan.
- Life-omics Research Division, Institute for Open and Transdisciplinary Research Initiative, Osaka University, Osaka, Japan.
- Laboratory of Bioimaging and Drug Discovery, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.
| |
Collapse
|
28
|
Yuan Y, Zhang Y, Lu X, Li J, Wang M, Zhang W, Zheng M, Sun Z, Xing Y, Li Y, Qu Y, Jiao Y, Han H, Xie C, Mao T. Novel insights into macrophage immunometabolism in nonalcoholic steatohepatitis. Int Immunopharmacol 2024; 131:111833. [PMID: 38503012 DOI: 10.1016/j.intimp.2024.111833] [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: 02/29/2024] [Accepted: 03/07/2024] [Indexed: 03/21/2024]
Abstract
Nonalcoholic steatohepatitis (NASH), an inflammatory subtype of nonalcoholic fatty liver disease (NAFLD), is characterized by liver steatosis, inflammation, hepatocellular injury and different degrees of fibrosis, and has been becoming the leading cause of liver-related morbidity and mortality worldwide. Unfortunately, the pathogenesis of NASH has not been completely clarified, and there are no approved therapeutic drugs. Recent accumulated evidences have revealed the involvement of macrophage in the regulation of host liver steatosis, inflammation and fibrosis, and different phenotypes of macrophages have different metabolic characteristics. Therefore, targeted regulation of macrophage immunometabolism may contribute to the treatment and prognosis of NASH. In this review, we summarized the current evidences of the role of macrophage immunometabolism in NASH, especially focused on the related function conversion, as well as the strategies to promote its polarization balance in the liver, and hold promise for macrophage immunometabolism-targeted therapies in the treatment of NASH.
Collapse
Affiliation(s)
- Yali Yuan
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Ye Zhang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Xinyu Lu
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Junxiang Li
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Muyuan Wang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Wenji Zhang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | | | | | - Yunqi Xing
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Yitong Li
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Yingdi Qu
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Yao Jiao
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Haixiao Han
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China.
| | - Chune Xie
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China; Shenzhen Bao'an Traditional Chinese Medicine Hospital, Shenzhen, PR China.
| | - Tangyou Mao
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China.
| |
Collapse
|
29
|
Jin J, Wang Y, Liu Y, Chakrabarti S, Su Z. Cardiac resident macrophages: Spatiotemporal distribution, development, physiological functions, and their translational potential on cardiac diseases. Acta Pharm Sin B 2024; 14:1483-1493. [PMID: 38572111 PMCID: PMC10985034 DOI: 10.1016/j.apsb.2023.12.018] [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: 10/10/2023] [Revised: 11/27/2023] [Accepted: 12/29/2023] [Indexed: 04/05/2024] Open
Abstract
Cardiac resident macrophages (CRMs) are the main population of cardiac immune cells. The role of these cells in regeneration, functional remodeling, and repair after cardiac injury is always the focus of research. However, in recent years, their dynamic changes and contributions in physiological states have a significant attention. CRMs have specific phenotypes and functions in different cardiac chambers or locations of the heart and at different stages. They further show specific differentiation and development processes. The present review will summarize the new progress about the spatiotemporal distribution, potential developmental regulation, and their roles in cardiac development and aging as well as the translational potential of CRMs on cardiac diseases. Of course, the research tools for CRMs, their respective advantages and disadvantages, and key issues on CRMs will further be discussed.
Collapse
Affiliation(s)
- Jing Jin
- International Genome Center, Jiangsu University, Zhenjiang 212013, China
- Institute for Medical Immunology, Jiangsu University, Zhenjiang 212013, China
| | - Yurou Wang
- International Genome Center, Jiangsu University, Zhenjiang 212013, China
- Institute for Medical Immunology, Jiangsu University, Zhenjiang 212013, China
| | - Yueqin Liu
- Center Laboratory, the Fourth People's Hospital of Zhenjiang, Zhenjiang 212008, China
| | - Subrata Chakrabarti
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario N6A 5C1, Canada
| | - Zhaoliang Su
- International Genome Center, Jiangsu University, Zhenjiang 212013, China
- Institute for Medical Immunology, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
30
|
Wöhner M, Brechtelsbauer S, Friedrich N, Vorsatz C, Bulang J, Liang C, Schorr L, Beschin A, Guilliams M, Ravetch J, Nimmerjahn F, Biburger M. Tissue niche occupancy determines the contribution of fetal- versus bone-marrow-derived macrophages to IgG effector functions. Cell Rep 2024; 43:113757. [PMID: 38354088 DOI: 10.1016/j.celrep.2024.113757] [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: 07/12/2023] [Revised: 12/20/2023] [Accepted: 01/23/2024] [Indexed: 02/16/2024] Open
Abstract
Understanding the mechanisms underlying cytotoxic immunoglobulin G (IgG) activity is critical for improving therapeutic antibody activity and inhibiting autoantibody-mediated tissue pathology. While prior research highlights the important role of the mononuclear phagocytic system for removing opsonized target cells, it remains unclear which monocyte or macrophage subsets stemming from fetal or post-natal bone-marrow (BM)-associated definitive hematopoiesis are involved in target cell depletion. By using a titrated irradiation approach as well as Kupffer-cell-specific deletion of activated Fcγ receptor signaling, we establish conditions under which the contribution of BM-derived monocytes versus yolk-sac-derived liver-resident macrophages to cytotoxic IgG activity can be studied. Our results demonstrate that liver-resident macrophages originating from either fetal or adult hematopoiesis play a central role in IgG-mediated depletion of opsonized target cells from the peripheral blood under steady-state conditions, highlighting the impact of the tissue niche and not macrophage origin for cytotoxic antibody activity.
Collapse
Affiliation(s)
- Miriam Wöhner
- Department of Biology, Division of Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Sarah Brechtelsbauer
- Department of Biology, Division of Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Niklas Friedrich
- Department of Biology, Division of Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Christof Vorsatz
- Department of Biology, Division of Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Johanna Bulang
- Department of Biology, Division of Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Chunguang Liang
- Institute of Immunology, University Hospital Jena, Leutragraben 3, 07743 Jena, Germany; Department of Bioinformatics, University of Würzburg, 97074 Würzburg, Germany
| | - Lena Schorr
- Department of Biology, Division of Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Alain Beschin
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1090 Brussels, Belgium; Myeloid Cell Immunology Laboratory, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Martin Guilliams
- Department of Biomedical Molecular Biology, Faculty of Science, Ghent University, 9000 Ghent, Belgium; Laboratory of Myeloid Cell Biology in Tissue Homeostasis and Regeneration, VIB-UGent Center for Inflammation Research, 9000 Ghent, Belgium
| | - Jeffrey Ravetch
- Laboratory of Molecular Genetics & Immunology, The Rockefeller University, New York, NY, USA
| | - Falk Nimmerjahn
- Department of Biology, Division of Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany; FAU Profile Center Immunomedicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany.
| | - Markus Biburger
- Department of Biology, Division of Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany; FAU Profile Center Immunomedicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany.
| |
Collapse
|
31
|
Krishnan A, Ozturk NB, Cutshaw KA, Guicciardi ME, Kitagataya T, Olson KE, Pavelko KD, Sherman W, Wixom AQ, Jalan-Sakrikar N, Baez-Faria M, Gutierrez F, Gores GJ. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) deletion in myeloid cells augments cholestatic liver injury. Sci Rep 2024; 14:2145. [PMID: 38273071 PMCID: PMC10810846 DOI: 10.1038/s41598-024-52710-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 01/23/2024] [Indexed: 01/27/2024] Open
Abstract
Ductular reactive (DR) cells exacerbate cholestatic liver injury and fibrosis. Herein, we posit that tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) emanates from recruited macrophages and restrains DR cell expansion, thereby limiting cholestatic liver injury. Wild type (WT), Trailfl/fl and myeloid-specific Trail deleted (TrailΔmye) C57BL/6 mice were exposed to DDC diet-induced cholestatic liver injury, which induced hepatomegaly and liver injury as compared to control diet-fed mice. However, parameters of liver injury, fibrosis, and inflammation were all increased in the TrailΔmye mice as compared to the WT and Trailfl/fl mice. High dimensional mass cytometry indicated that cholestasis resulted in increased hepatic recruitment of subsets of macrophages and neutrophils in the TrailΔmye mice. Spatial transcriptomics analysis revealed that the PanCK+ cholangiocytes from TrailΔmye mice had increased expression of the known myeloid attractants S100a8, Cxcl5, Cx3cl1, and Cxcl1. Additionally, in situ hybridization of Cxcl1, a potent neutrophil chemoattractant, demonstrated an increased expression in CK19+ cholangiocytes of TrailΔmye mice. Collectively, these data suggest that TRAIL from myeloid cells, particularly macrophages, restrains a subset of DR cells (i.e., Cxcl1 positive cells), limiting liver inflammation and fibrosis. Reprogramming macrophages to express TRAIL may be salutary in cholestasis.
Collapse
Affiliation(s)
- Anuradha Krishnan
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Nazli Begum Ozturk
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Kaiyel A Cutshaw
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Maria Eugenia Guicciardi
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Takashi Kitagataya
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Kirsta E Olson
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | | | - William Sherman
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Alexander Q Wixom
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Nidhi Jalan-Sakrikar
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Michelle Baez-Faria
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Florencia Gutierrez
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Gregory J Gores
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
32
|
Chen G, Hu X, Huang Y, Xiang X, Pan S, Chen R, Xu X. Role of the immune system in liver transplantation and its implications for therapeutic interventions. MedComm (Beijing) 2023; 4:e444. [PMID: 38098611 PMCID: PMC10719430 DOI: 10.1002/mco2.444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 12/17/2023] Open
Abstract
Liver transplantation (LT) stands as the gold standard for treating end-stage liver disease and hepatocellular carcinoma, yet postoperative complications continue to impact survival rates. The liver's unique immune system, governed by a microenvironment of diverse immune cells, is disrupted during processes like ischemia-reperfusion injury posttransplantation, leading to immune imbalance, inflammation, and subsequent complications. In the posttransplantation period, immune cells within the liver collaboratively foster a tolerant environment, crucial for immune tolerance and liver regeneration. While clinical trials exploring cell therapy for LT complications exist, a comprehensive summary is lacking. This review provides an insight into the intricacies of the liver's immune microenvironment, with a specific focus on macrophages and T cells as primary immune players. Delving into the immunological dynamics at different stages of LT, we explore the disruptions after LT and subsequent immune responses. Focusing on immune cell targeting for treating liver transplant complications, we provide a comprehensive summary of ongoing clinical trials in this domain, especially cell therapies. Furthermore, we offer innovative treatment strategies that leverage the opportunities and prospects identified in the therapeutic landscape. This review seeks to advance our understanding of LT immunology and steer the development of precise therapies for postoperative complications.
Collapse
Affiliation(s)
- Guanrong Chen
- The Fourth School of Clinical MedicineZhejiang Chinese Medical UniversityHangzhouChina
| | - Xin Hu
- Zhejiang University School of MedicineHangzhouChina
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang ProvinceHangzhouChina
| | - Yingchen Huang
- The Fourth School of Clinical MedicineZhejiang Chinese Medical UniversityHangzhouChina
| | - Xiaonan Xiang
- Zhejiang University School of MedicineHangzhouChina
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang ProvinceHangzhouChina
| | - Sheng Pan
- Zhejiang University School of MedicineHangzhouChina
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang ProvinceHangzhouChina
| | - Ronggao Chen
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Xiao Xu
- Zhejiang University School of MedicineHangzhouChina
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang ProvinceHangzhouChina
- Zhejiang Chinese Medical UniversityHangzhouChina
| |
Collapse
|
33
|
Fa P, Ke BG, Dupre A, Tsung A, Zhang H. The implication of neutrophil extracellular traps in nonalcoholic fatty liver disease. Front Immunol 2023; 14:1292679. [PMID: 38022519 PMCID: PMC10652891 DOI: 10.3389/fimmu.2023.1292679] [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: 09/11/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is an expanding worldwide health concern, and the underlying mechanisms contributing to its progression still need further exploration. Neutrophil extracellular traps (NETs) are intricate formations comprised of nuclear constituents and diverse antimicrobial granules that are released into the extracellular milieu by activated neutrophils upon various triggers, which play a pivotal part in the onset and advancement of NAFLD. NETs actively participate in the genesis of NAFLD by fostering oxidative stress and inflammation, ultimately resulting in hepatic fat accumulation and the escalation of liver injury. Recent insights into the interaction with other hepatic immune populations and mediators, such as macrophages and T regulatory cells, have revealed several important mechanisms that can trigger further liver injury. In conclusion, the formation of NETs emerged as an important factor in the development of NAFLD, offering a promising target for innovative therapeutic approaches against this debilitating condition. This comprehensive review seeks to compile existing studies exploring the involvement of NETs in the genesis of NAFLD and their influence on the immune response throughout the progression of NAFLD.
Collapse
Affiliation(s)
- Pengyan Fa
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Benjamin G. Ke
- School of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Abigail Dupre
- School of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Allan Tsung
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Hongji Zhang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
34
|
Han M, Geng J, Zhang S, Rao J, Zhu Y, Xu S, Wang F, Ma F, Zhou M, Zhou H. Invariant natural killer T cells drive hepatic homeostasis in nonalcoholic fatty liver disease via sustained IL-10 expression in CD170 + Kupffer cells. Eur J Immunol 2023; 53:e2350474. [PMID: 37489253 DOI: 10.1002/eji.202350474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 07/05/2023] [Accepted: 07/24/2023] [Indexed: 07/26/2023]
Abstract
Kupffer cells (KCs) are liver-resident macrophages involved in hepatic inflammatory responses, including nonalcoholic fatty liver disease (NAFLD) development. However, the contribution of KC subsets to liver inflammation remains unclear. Here, using high-dimensional single-cell RNA sequencing, we characterized murine embryo-derived KCs and identified two KC populations with different gene expression profiles: KC-1 and KC-2. KC-1 expressed CD170, exhibiting immunoreactivity and immune-regulatory abilities, while KC-2 highly expressed lipid metabolism-associated genes. In a high-fat diet-induced NAFLD model, KC-1 cells differentiated into pro-inflammatory phenotypes and initiated more frequent communications with invariant natural killer T (iNKT) cells. In KC-1, interleukin (IL)-10 expression was unaffected by the high-fat diet but impaired by iNKT cell ablation and upregulated by iNKT cell adoptive transfer in vivo. Moreover, in a cellular co-culture system, primary hepatic iNKT cells promoted IL-10 expression in RAW264.7 and primary KC-1 cells. CD206 signal blocking in KC-1 or CD206 knockdown in RAW264.7 cells significantly reduced IL-10 expression. In conclusion, we identified two embryo-derived KC subpopulations with distinct transcriptional profiles. The CD206-mediated crosstalk between iNKT and KC-1 cells maintains IL-10 expression in KC-1 cells, affecting hepatic immune balance. Therefore, KC-based therapeutic strategies must consider cellular heterogeneity and the local immune microenvironment for enhanced specificity and efficiency.
Collapse
Affiliation(s)
- Mutian Han
- Department of Immunology, College of Basic Medical Science, Anhui Medical University, Anhui, China
| | - Jinke Geng
- Department of Immunology, College of Basic Medical Science, Anhui Medical University, Anhui, China
| | - Shuangshuang Zhang
- Department of Immunology, College of Basic Medical Science, Anhui Medical University, Anhui, China
| | - Jia Rao
- Department of Immunology, College of Basic Medical Science, Anhui Medical University, Anhui, China
| | - Yansong Zhu
- Department of Cell and Biology, College of Life Sciences, Anhui Medical University, Anhui, China
| | - Shaodong Xu
- Department of Cell and Biology, College of Life Sciences, Anhui Medical University, Anhui, China
| | - Fei Wang
- Department of Immunology, College of Basic Medical Science, Anhui Medical University, Anhui, China
| | - Fang Ma
- Center for Scientific Research, Anhui Medical University, Anhui, China
| | - Meng Zhou
- Department of Cell and Biology, College of Life Sciences, Anhui Medical University, Anhui, China
| | - Hong Zhou
- Department of Immunology, College of Basic Medical Science, Anhui Medical University, Anhui, China
- Department of Cell and Biology, College of Life Sciences, Anhui Medical University, Anhui, China
| |
Collapse
|
35
|
Ruiqi W, Xiaoli F, Leyu Z, Mengyi S, Qiaoyu D, Yanyi Z, Li Y. Monocyte-derived macrophages contribute to the deterioration of immunological liver injury in mice. Int Immunopharmacol 2023; 124:111036. [PMID: 37832236 DOI: 10.1016/j.intimp.2023.111036] [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: 06/16/2023] [Revised: 09/28/2023] [Accepted: 10/05/2023] [Indexed: 10/15/2023]
Abstract
BACKGROUND & AIMS Autoimmune hepatitis (AIH) is characterized by hepatocyte destruction, leading to lymphocyte and macrophage accumulation in the liver. However, the specific mechanisms of how macrophages participate in the occurrence and development of AIH are still unclear. In this study, we investigated the effect of monocyte-derived macrophages on Con A-induced immunological liver injury in mice and we hypothesized that inhibition of CCR2 with the dual CCR2/5 inhibitor, cenicriviroc (CVC), would attenuate Con A-induced hepatitis in mice by reducing the recruitment of monocytes into the liver. METHODS Murine experimental AIH was established by concanavalin A (Con A) injection intravenously. Macrophages were depleted by injection of clodronate liposomes in Con A-treated mice. Moreover, inhibition of the CCR2/5 signaling pathway in Con A mice is achieved by CVC. Liver injury and infiltration of monocyte-derived macrophages were assessed by serum transaminase levels, histopathology, immunohistochemistry, flow cytometry, RT-qPCR, ELISA, TUNEL assay and dihydroethidium staining. RESULTS The number of macrophages in the mouse livers increased in the Con A-induced hepatitis mouse model, and flow cytometry showed a significant increase in the proportion of F4/80loCD11bhi monocyte-derived macrophages, while there was no significant change in the proportion of F4/80hiCD11blo Kupffer cells. After the depletion of liver macrophages by clodronate liposomes, the levels of serum ALT and AST, and the degree of liver tissue damage were alleviated in Con A-treated mice. Furthermore, Con A leaded an increase in the expression of a group of CC chemokines in mouse livers, and the elevation of CCL2 was prevented with the depletion of macrophages. Additionally, CVC reduced macrophage infiltration in the liver and ameliorated Con A-induced liver injury. Meanwhile, CVC reduced the apoptosis and oxidative damage of hepatocytes caused by Con A. CONCLUSIONS Our research demonstrates that there is an increase in monocyte-derived macrophages in the livers due to the monocyte infiltration resulted from the activation of the CCL2-CCR2 axis in Con A-induced liver injury mouse model. Pharmacological inhibition of CCR2 monocyte recruitment by CVC efficiently ameliorates the hepatic inflammation, indicating the therapeutic potential of CVC in patients with AIH.
Collapse
Affiliation(s)
- Wu Ruiqi
- Department of Gastroenterology and Hepatology and Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fan Xiaoli
- Department of Gastroenterology and Hepatology and Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhou Leyu
- Department of Gastroenterology and Hepatology and Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shen Mengyi
- Department of Gastroenterology and Hepatology and Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Deng Qiaoyu
- Department of Gastroenterology and Hepatology and Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zheng Yanyi
- Department of Gastroenterology and Hepatology and Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yang Li
- Department of Gastroenterology and Hepatology and Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
36
|
Li W, Yang Y, Yang L, Chang N, Li L. Monocyte-derived Kupffer cells dominate in the Kupffer cell pool during liver injury. Cell Rep 2023; 42:113164. [PMID: 37740916 DOI: 10.1016/j.celrep.2023.113164] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 06/12/2023] [Accepted: 09/07/2023] [Indexed: 09/25/2023] Open
Abstract
Healthy Kupffer cell (KC) pool is dominated by embryonic KCs (EmKCs), preserving liver homeostasis. How the KC pool varies upon injury remains unclear. Using chimeric mice with bone marrow (BM) cells labeled with enhanced green fluorescent protein, we identify that BM monocyte-derived KCs (MoKCs) become dominant in cholestatic- or toxic-injured livers via immunofluorescence and mass cytometry. Single-cell RNA sequencing (scRNA-seq) unveils the enhanced proliferative, anti-apoptotic properties and repair potential of MoKCs compared with EmKCs, which are confirmed in vivo and ex vivo through flow cytometry, qPCR, Cell Counting Kit-8, and immunofluorescence. Furthermore, compared with EmKC-dominated livers, MoKC-dominated livers exhibit less functional damage, necrosis, and fibrosis under damage, as tested by serum alanine aminotransferase activity detection, H&E and Sirius red staining, qPCR, and western blot. Collectively, we highlight that MoKCs dominate the KC pool in injured livers and show enhanced proliferative and anti-apoptotic properties while also promoting repair and attenuating fibrosis.
Collapse
Affiliation(s)
- Weiyang Li
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing 100069, China
| | - Yuanru Yang
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing 100069, China
| | - Lin Yang
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing 100069, China
| | - Na Chang
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing 100069, China.
| | - Liying Li
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
37
|
Xie D, Ouyang S. The role and mechanisms of macrophage polarization and hepatocyte pyroptosis in acute liver failure. Front Immunol 2023; 14:1279264. [PMID: 37954583 PMCID: PMC10639160 DOI: 10.3389/fimmu.2023.1279264] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/16/2023] [Indexed: 11/14/2023] Open
Abstract
Acute liver failure (ALF) is a severe liver disease caused by disruptions in the body's immune microenvironment. In the early stages of ALF, Kupffer cells (KCs) become depleted and recruit monocytes derived from the bone marrow or abdomen to replace the depleted macrophages entering the liver. These monocytes differentiate into mature macrophages, which are activated in the immune microenvironment of the liver and polarized to perform various functions. Macrophage polarization can occur in two directions: pro-inflammatory M1 macrophages and anti-inflammatory M2 macrophages. Controlling the ratio and direction of M1 and M2 in ALF can help reduce liver injury. However, the liver damage caused by pyroptosis should not be underestimated, as it is a caspase-dependent form of cell death. Inhibiting pyroptosis has been shown to effectively reduce liver damage induced by ALF. Furthermore, macrophage polarization and pyroptosis share common binding sites, signaling pathways, and outcomes. In the review, we describe the role of macrophage polarization and pyroptosis in the pathogenesis of ALF. Additionally, we preliminarily explore the relationship between macrophage polarization and pyroptosis, as well as their effects on ALF.
Collapse
Affiliation(s)
| | - Shi Ouyang
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, Department of Infectious Diseases, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
38
|
Yamaji K, Iwabuchi S, Tokunaga Y, Hashimoto S, Yamane D, Toyama S, Kono R, Kitab B, Tsukiyama-Kohara K, Osawa Y, Hayashi Y, Hishima T, Tateno C, Kimura K, Okanoue T, Kohara M. Molecular insights of a CBP/β-catenin-signaling inhibitor on nonalcoholic steatohepatitis-induced liver fibrosis and disorder. Biomed Pharmacother 2023; 166:115379. [PMID: 37647690 DOI: 10.1016/j.biopha.2023.115379] [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: 06/16/2023] [Revised: 08/04/2023] [Accepted: 08/23/2023] [Indexed: 09/01/2023] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is a progressive fibrotic disease associated with an increased risk of developing hepatocellular carcinoma; at present, no efficient therapeutic strategy has been established. Herein, we examined the efficacy of PRI-724, a potent inhibitor of CBP/β-catenin signaling, for treating NASH-related liver fibrosis and disorder and characterized its mechanism. Choline-deficient, L-amino acid-defined, high-fat diet (CDAHFD)-fed mice exhibited NASH-induced liver fibrosis that is characterized by steatosis, lobular inflammation, hepatocellular injury and collagen fibrils. To examine the therapeutic effect, CDAHFD-fed mice were administered PRI-724. Serum levels of ALT and pro-fibrotic molecule, i.e. Mac-2 bp, alpha smooth muscle actin, type I and type III collagens, decreased significantly. mRNA levels of the matrix metalloproteinases Mmp8 and Mmp9 in the liver were significantly increased, and increases in the abundance of MMP9-producing neutrophils and macrophages were observed. Marco+Mmp9+Cd68+ Kupffer cells were only observed in the livers of mice treated with PRI-724, and Mmp9 expression in Marco+Cd68+ Kupffer cells increased 4.3-fold. Moreover, hepatic expression of the lipid metabolism regulator, pyruvate dehydrogenase kinase 4 and liver lipid droplets also decreased significantly. PRI-724-treated NASH mice not only recovered from NASH-related liver fibrosis through the effect of PRI-724 down-regulating the expression of pro-fibrotic genes and up-regulating the expression of anti-fibrotic genes, but they also recovered from NASH-induced liver disorder. PRI-724, a selective CBP/β-catenin inhibitor, thus shows a potent therapeutic effect for NASH-related liver fibrosis and for decreasing adipose tissue in the liver.
Collapse
Affiliation(s)
- Kenzaburo Yamaji
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Sadahiro Iwabuchi
- Department of Molecular Pathophysiology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama 641-8509, Japan
| | - Yuko Tokunaga
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Shinichi Hashimoto
- Department of Molecular Pathophysiology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama 641-8509, Japan
| | - Daisuke Yamane
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Sakiko Toyama
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Risa Kono
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Bouchra Kitab
- Transboundary Animal Diseases Center, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan
| | - Kyoko Tsukiyama-Kohara
- Transboundary Animal Diseases Center, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan.
| | - Yosuke Osawa
- Department of Gastroenterology, International University of Health and Welfare Hospital, Nasushiobara 324-8501, Japan
| | - Yukiko Hayashi
- Department of Pathology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo 113-8677, Japan
| | - Tsunekazu Hishima
- Department of Pathology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo 113-8677, Japan
| | - Chise Tateno
- R&D Department, PhoenixBio Co., Ltd., 3-4-1 Kagamiyama, Higashihiroshima, Hiroshima 739-0046, Japan
| | - Kiminori Kimura
- Department of Hepatology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo 113-8677, Japan
| | - Takeshi Okanoue
- Department of Gastroenterology and Hepatology, Saiseikai Suita Hospital, Osaka 564-0013, Japan
| | - Michinori Kohara
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan.
| |
Collapse
|
39
|
Makiuchi N, Takano S, Tada Y, Kasai K, Igarashi N, Kani K, Kato M, Goto K, Matsuura Y, Ichimura-Shimizu M, Furusawa Y, Tsuneyama K, Nagai Y. Dynamics of Liver Macrophage Subsets in a Novel Mouse Model of Non-Alcoholic Steatohepatitis Using C57BL/6 Mice. Biomedicines 2023; 11:2659. [PMID: 37893033 PMCID: PMC10604124 DOI: 10.3390/biomedicines11102659] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/26/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
Macrophages are critical for the development of non-alcoholic steatohepatitis (NASH). Our previous findings in TSNO mouse livers showed that an iHFC (high-fat/cholesterol/cholate) diet induced liver fibrosis similar to human NASH and led to the accumulation of distinct subsets of macrophage: CD11c+/Ly6C- and CD11c-/Ly6C+ cells. CD11c+/Ly6C- cells were associated with the promotion of advanced liver fibrosis in NASH. On the other hand, CD11c-/Ly6C+ cells exhibited an anti-inflammatory effect and were involved in tissue remodeling processes. This study aimed to elucidate whether an iHFC diet with reduced cholic acid (iHFC#2 diet) induces NASH in C57BL/6 mice and examine the macrophage subsets accumulating in the liver. Histological and quantitative real-time PCR analyses revealed that the iHFC#2 diet promoted inflammation and fibrosis indicative of NASH in the livers of C57BL/6 mice. Cell numbers of Kupffer cells decreased and recruited macrophages were accumulated in the livers of iHFC#2 diet-fed C57BL/6 mice. Notably, the iHFC#2 diet resulted in the accumulation of three macrophage subsets in the livers of C57BL/6 mice: CD11c+/Ly6C-, CD11c-/Ly6C+, and CD11c+/Ly6C+ cells. However, CD11c+/Ly6C+ cells were not distinct populations in the iHFC-fed TSNO mice. Thus, differences in cholic acid content and mouse strain affect the macrophage subsets that accumulate in the liver.
Collapse
Affiliation(s)
- Nana Makiuchi
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan; (N.M.); (S.T.); (Y.T.); (K.K.); (N.I.); (K.K.); (M.K.); (K.G.); (Y.M.); (Y.F.)
| | - Shun Takano
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan; (N.M.); (S.T.); (Y.T.); (K.K.); (N.I.); (K.K.); (M.K.); (K.G.); (Y.M.); (Y.F.)
| | - Yuki Tada
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan; (N.M.); (S.T.); (Y.T.); (K.K.); (N.I.); (K.K.); (M.K.); (K.G.); (Y.M.); (Y.F.)
| | - Kaichi Kasai
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan; (N.M.); (S.T.); (Y.T.); (K.K.); (N.I.); (K.K.); (M.K.); (K.G.); (Y.M.); (Y.F.)
| | - Naoya Igarashi
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan; (N.M.); (S.T.); (Y.T.); (K.K.); (N.I.); (K.K.); (M.K.); (K.G.); (Y.M.); (Y.F.)
| | - Koudai Kani
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan; (N.M.); (S.T.); (Y.T.); (K.K.); (N.I.); (K.K.); (M.K.); (K.G.); (Y.M.); (Y.F.)
| | - Miyuna Kato
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan; (N.M.); (S.T.); (Y.T.); (K.K.); (N.I.); (K.K.); (M.K.); (K.G.); (Y.M.); (Y.F.)
| | - Kana Goto
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan; (N.M.); (S.T.); (Y.T.); (K.K.); (N.I.); (K.K.); (M.K.); (K.G.); (Y.M.); (Y.F.)
| | - Yudai Matsuura
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan; (N.M.); (S.T.); (Y.T.); (K.K.); (N.I.); (K.K.); (M.K.); (K.G.); (Y.M.); (Y.F.)
| | - Mayuko Ichimura-Shimizu
- Department of Pathology and Laboratory Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-8-15 Kuramoto-cho, Tokushima 770-8503, Japan; (M.I.-S.); (K.T.)
| | - Yukihiro Furusawa
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan; (N.M.); (S.T.); (Y.T.); (K.K.); (N.I.); (K.K.); (M.K.); (K.G.); (Y.M.); (Y.F.)
| | - Koichi Tsuneyama
- Department of Pathology and Laboratory Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-8-15 Kuramoto-cho, Tokushima 770-8503, Japan; (M.I.-S.); (K.T.)
| | - Yoshinori Nagai
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan; (N.M.); (S.T.); (Y.T.); (K.K.); (N.I.); (K.K.); (M.K.); (K.G.); (Y.M.); (Y.F.)
| |
Collapse
|
40
|
Hassan GS, Flores Molina M, Shoukry NH. The multifaceted role of macrophages during acute liver injury. Front Immunol 2023; 14:1237042. [PMID: 37736102 PMCID: PMC10510203 DOI: 10.3389/fimmu.2023.1237042] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/15/2023] [Indexed: 09/23/2023] Open
Abstract
The liver is situated at the interface of the gut and circulation where it acts as a filter for blood-borne and gut-derived microbes and biological molecules, promoting tolerance of non-invasive antigens while driving immune responses against pathogenic ones. Liver resident immune cells such as Kupffer cells (KCs), a subset of macrophages, maintain homeostasis under physiological conditions. However, upon liver injury, these cells and others recruited from circulation participate in the response to injury and the repair of tissue damage. Such response is thus spatially and temporally regulated and implicates interconnected cells of immune and non-immune nature. This review will describe the hepatic immune environment during acute liver injury and the subsequent wound healing process. In its early stages, the wound healing immune response involves a necroinflammatory process characterized by partial depletion of resident KCs and lymphocytes and a significant infiltration of myeloid cells including monocyte-derived macrophages (MoMFs) complemented by a wave of pro-inflammatory mediators. The subsequent repair stage includes restoring KCs, initiating angiogenesis, renewing extracellular matrix and enhancing proliferation/activation of resident parenchymal and mesenchymal cells. This review will focus on the multifaceted role of hepatic macrophages, including KCs and MoMFs, and their spatial distribution and roles during acute liver injury.
Collapse
Affiliation(s)
- Ghada S. Hassan
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Manuel Flores Molina
- 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
| | - 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
|
41
|
Musrati MA, De Baetselier P, Movahedi K, Van Ginderachter JA. Ontogeny, functions and reprogramming of Kupffer cells upon infectious disease. Front Immunol 2023; 14:1238452. [PMID: 37691953 PMCID: PMC10485603 DOI: 10.3389/fimmu.2023.1238452] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 08/11/2023] [Indexed: 09/12/2023] Open
Abstract
The liver is a vital metabolic organ that also performs important immune-regulatory functions. In the context of infections, the liver represents a target site for various pathogens, while also having an outstanding capacity to filter the blood from pathogens and to contain infections. Pathogen scavenging by the liver is primarily performed by its large and heterogeneous macrophage population. The major liver-resident macrophage population is located within the hepatic microcirculation and is known as Kupffer cells (KCs). Although other minor macrophages reside in the liver as well, KCs remain the best characterized and are the best well-known hepatic macrophage population to be functionally involved in the clearance of infections. The response of KCs to pathogenic insults often governs the overall severity and outcome of infections on the host. Moreover, infections also impart long-lasting, and rarely studied changes to the KC pool. In this review, we discuss current knowledge on the biology and the various roles of liver macrophages during infections. In addition, we reflect on the potential of infection history to imprint long-lasting effects on macrophages, in particular liver macrophages.
Collapse
Affiliation(s)
- Mohamed Amer Musrati
- Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Patrick De Baetselier
- Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Kiavash Movahedi
- Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
- Lab of Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jo A. Van Ginderachter
- Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| |
Collapse
|
42
|
Park HJ, Choi J, Kim H, Yang DY, An TH, Lee EW, Han BS, Lee SC, Kim WK, Bae KH, Oh KJ. Cellular heterogeneity and plasticity during NAFLD progression. Front Mol Biosci 2023; 10:1221669. [PMID: 37635938 PMCID: PMC10450943 DOI: 10.3389/fmolb.2023.1221669] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/18/2023] [Indexed: 08/29/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a progressive liver disease that can progress to nonalcoholic steatohepatitis (NASH), NASH-related cirrhosis, and hepatocellular carcinoma (HCC). NAFLD ranges from simple steatosis (or nonalcoholic fatty liver [NAFL]) to NASH as a progressive form of NAFL, which is characterized by steatosis, lobular inflammation, and hepatocellular ballooning with or without fibrosis. Because of the complex pathophysiological mechanism and the heterogeneity of NAFLD, including its wide spectrum of clinical and histological characteristics, no specific therapeutic drugs have been approved for NAFLD. The heterogeneity of NAFLD is closely associated with cellular plasticity, which describes the ability of cells to acquire new identities or change their phenotypes in response to environmental stimuli. The liver consists of parenchymal cells including hepatocytes and cholangiocytes and nonparenchymal cells including Kupffer cells, hepatic stellate cells, and endothelial cells, all of which have specialized functions. This heterogeneous cell population has cellular plasticity to adapt to environmental changes. During NAFLD progression, these cells can exert diverse and complex responses at multiple levels following exposure to a variety of stimuli, including fatty acids, inflammation, and oxidative stress. Therefore, this review provides insights into NAFLD heterogeneity by addressing the cellular plasticity and metabolic adaptation of hepatocytes, cholangiocytes, hepatic stellate cells, and Kupffer cells during NAFLD progression.
Collapse
Affiliation(s)
- Hyun-Ju Park
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Juyong Choi
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Hyunmi Kim
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Da-Yeon Yang
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Tae Hyeon An
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Eun-Woo Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Baek-Soo Han
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
- Biodefense Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Sang Chul Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Won Kon Kim
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Kwang-Hee Bae
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Kyoung-Jin Oh
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| |
Collapse
|
43
|
Li X, Huai Q, Zhu C, Zhang X, Xu W, Dai H, Wang H. GDF15 Ameliorates Liver Fibrosis by Metabolic Reprogramming of Macrophages to Acquire Anti-Inflammatory Properties. Cell Mol Gastroenterol Hepatol 2023; 16:711-734. [PMID: 37499753 PMCID: PMC10520366 DOI: 10.1016/j.jcmgh.2023.07.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/19/2023] [Accepted: 07/19/2023] [Indexed: 07/29/2023]
Abstract
BACKGROUND & AIMS Liver fibrosis/cirrhosis is significant health burden worldwide, resulting in liver failure or cancer and accounting for many deaths each year. The pathogenesis of liver fibrosis is very complex, which makes treatment challenging. Growth differentiation factor 15 (GDF15), a cysteine knot protein belonging to the transforming growth factor β (TGF-β) superfamily, has been shown to play a protective role after tissue injury and to promote a negative energy balance during obesity and diabetes. However, paucity of literature is available about GDF15 function in liver fibrosis. This study aimed to investigate the immunomodulatory role and therapeutic potential of GDF15 in progression of hepatic fibrosis. METHODS GDF15 expression was studied in patients with fibrosis/cirrhosis and in 2 murine models of liver fibrosis, including mice treated with CCl4 or DDC diet. GDF15 involvement in the pathogenesis of liver fibrosis was assessed in Gdf15 knockout mouse using both CCl4 and DDC diet experimental models. We used the CCl4 and/or DDC diet-induced liver fibrosis model to examine the antifibrotic and anti-inflammatory effects of AAV8-mediated GDF15 overexpression in hepatocytes or recombinant mouse GDF15. RESULTS GDF15 expression is decreased in the liver of animal models and patients with liver fibrosis/cirrhosis compared with those without liver disease. In vivo studies showed that GDF15 deficiency aggravated CCl4 and DDC diet-induced liver fibrosis, while GDF15 overexpression mediated by AAV8 or its recombinant protein alleviated CCl4 and/or DDC diet-induced liver fibrosis. In Gdf15 knockout mice, the intrahepatic microenvironment that developed during fibrosis showed relatively more inflammation, as demonstrated by enhanced infiltration of monocytes and neutrophils and increased expression of proinflammatory factors, which could be diminished by AAV8-mediated GDF15 overexpression in hepatocytes. Intriguingly, GDF15 exerts its effects by reprogramming the metabolic pathways of macrophages to acquire an oxidative phosphorylation-dependent anti-inflammatory functional fate. Furthermore, adoptive transfer of GDF15-preprogrammed macrophages to mouse models of liver fibrosis induced by CCl4 attenuated inflammation and alleviated the progression of liver fibrosis. CONCLUSION GDF15 ameliorates liver fibrosis via modulation of liver macrophages. Our data implicate the importance of the liver microenvironment in macrophage programming during liver fibrosis and suggest that GDF15 is a potentially attractive therapeutic target for the treatment of patients with liver fibrosis.
Collapse
Affiliation(s)
- Xiaolei Li
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qian Huai
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Cheng Zhu
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xu Zhang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wentao Xu
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hanren Dai
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China.
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China.
| |
Collapse
|
44
|
He W, Huang Y, Shi X, Wang Q, Wu M, Li H, Liu Q, Zhang X, Huang C, Li X. Identifying a distinct fibrosis subset of NAFLD via molecular profiling and the involvement of profibrotic macrophages. J Transl Med 2023; 21:448. [PMID: 37415134 PMCID: PMC10326954 DOI: 10.1186/s12967-023-04300-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 06/23/2023] [Indexed: 07/08/2023] Open
Abstract
BACKGROUND There are emerging studies suggesting that non-alcoholic fatty liver disease (NAFLD) is a heterogeneous disease with multiple etiologies and molecular phenotypes. Fibrosis is the key process in NAFLD progression. In this study, we aimed to explore molecular phenotypes of NAFLD with a particular focus on the fibrosis phenotype and also aimed to explore the changes of macrophage subsets in the fibrosis subset of NAFLD. METHODS To assess the transcriptomic alterations of key factors in NAFLD and fibrosis progression, we included 14 different transcriptomic datasets of liver tissues. In addition, two single-cell RNA sequencing (scRNA-seq) datasets were included to construct transcriptomic signatures that could represent specific cells. To explore the molecular subsets of fibrosis in NAFLD based on the transcriptomic features, we used a high-quality RNA-sequencing (RNA-seq) dataset of liver tissues from patients with NAFLD. Non-negative matrix factorization (NMF) was used to analyze the molecular subsets of NAFLD based on the gene set variation analysis (GSVA) enrichment scores of key molecule features in liver tissues. RESULTS The key transcriptomic signatures on NAFLD including non-alcoholic steatohepatitis (NASH) signature, fibrosis signature, non-alcoholic fatty liver (NAFL) signature, liver aging signature and TGF-β signature were constructed by liver transcriptome datasets. We analyzed two liver scRNA-seq datasets and constructed cell type-specific transcriptomic signatures based on the genes that were highly expressed in each cell subset. We analyzed the molecular subsets of NAFLD by NMF and categorized four main subsets of NAFLD. Cluster 4 subset is mainly characterized by liver fibrosis. Patients with Cluster 4 subset have more advanced liver fibrosis than patients with other subsets, or may have a high risk of liver fibrosis progression. Furthermore, we identified two key monocyte-macrophage subsets which were both significantly correlated with the progression of liver fibrosis in NAFLD patients. CONCLUSION Our study revealed the molecular subtypes of NAFLD by integrating key information from transcriptomic expression profiling and liver microenvironment, and identified a novel and distinct fibrosis subset of NAFLD. The fibrosis subset is significantly correlated with the profibrotic macrophages and M2 macrophage subset. These two liver macrophage subsets may be important players in the progression of liver fibrosis of NAFLD patients.
Collapse
Affiliation(s)
- Weiwei He
- Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xaimen, China
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Translational Medicine for Diabetes, Xiamen, China
| | - Yinxiang Huang
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Translational Medicine for Diabetes, Xiamen, China
| | - Xiulin Shi
- Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xaimen, China
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Translational Medicine for Diabetes, Xiamen, China
| | - Qingxuan Wang
- Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xaimen, China
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Translational Medicine for Diabetes, Xiamen, China
| | - Menghua Wu
- Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xaimen, China
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Translational Medicine for Diabetes, Xiamen, China
| | - Han Li
- Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xaimen, China
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Translational Medicine for Diabetes, Xiamen, China
| | - Qiuhong Liu
- Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xaimen, China
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Translational Medicine for Diabetes, Xiamen, China
| | - Xiaofang Zhang
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Translational Medicine for Diabetes, Xiamen, China
| | - Caoxin Huang
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, China.
- Fujian Provincial Key Laboratory of Translational Medicine for Diabetes, Xiamen, China.
| | - Xuejun Li
- Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xaimen, China.
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, China.
- Fujian Provincial Key Laboratory of Translational Medicine for Diabetes, Xiamen, China.
| |
Collapse
|
45
|
Peng B, Li H, Liu K, Zhang P, Zhuang Q, Li J, Yang M, Cheng K, Ming Y. Intrahepatic macrophage reprogramming associated with lipid metabolism in hepatitis B virus-related acute-on-chronic liver failure. J Transl Med 2023; 21:419. [PMID: 37380987 DOI: 10.1186/s12967-023-04294-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 06/22/2023] [Indexed: 06/30/2023] Open
Abstract
BACKGROUND Acute-on-chronic liver failure (ACLF) is a severe syndrome with high short-term mortality, but the pathophysiology still remains largely unknown. Immune dysregulation and metabolic disorders contribute to the progression of ACLF, but the crosstalk between immunity and metabolism during ACLF is less understood. This study aims to depict the immune microenvironment in the liver during ACLF, and explore the role of lipid metabolic disorder on immunity. METHODS Single-cell RNA-sequencing (scRNA-seq) was performed using the liver non-parenchymal cells (NPCs) and peripheral blood mononuclear cells (PBMCs) from healthy controls, cirrhosis patients and ACLF patients. A series of inflammation-related cytokines and chemokines were detected using liver and plasma samples. The lipid metabolomics targeted free fatty acids (FFAs) in the liver was also detected. RESULTS The scRNA-seq analysis of liver NPCs showed a significant increase of monocytes/macrophages (Mono/Mac) infiltration in ACLF livers, whereas the resident Kupffer cells (KCs) were exhausted. A characterized TREM2+ Mono/Mac subpopulation was identified in ACLF, and showed immunosuppressive function. Combined with the scRNA-seq data from PBMCs, the pseudotime analysis revealed that the TREM2+ Mono/Mac were differentiated from the peripheral monocytes and correlated with lipid metabolism-related genes including APOE, APOC1, FABP5 and TREM2. The targeted lipid metabolomics proved the accumulation of unsaturated FFAs associated with α-linolenic acid (α-LA) and α-LA metabolism and beta oxidation of very long chain fatty acids in the ACLF livers, indicating that unsaturated FFAs might promote the differentiation of TREM2+ Mono/Mac during ACLF. CONCLUSIONS The reprogramming of macrophages was found in the liver during ACLF. The immunosuppressive TREM2+ macrophages were enriched in the ACLF liver and contributed to the immunosuppressive hepatic microenvironment. The accumulation of unsaturated FFAs in the ACLF liver promoted the reprogramming of the macrophages. It might be a potential target to improve the immune deficiency of ACLF patients through regulating lipid metabolism.
Collapse
Affiliation(s)
- Bo Peng
- Transplantation Center, The Third Xiangya Hospital, Central South University, Hunan, 410013, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Hao Li
- Transplantation Center, The Third Xiangya Hospital, Central South University, Hunan, 410013, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Kai Liu
- Transplantation Center, The Third Xiangya Hospital, Central South University, Hunan, 410013, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Pengpeng Zhang
- Transplantation Center, The Third Xiangya Hospital, Central South University, Hunan, 410013, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Quan Zhuang
- Transplantation Center, The Third Xiangya Hospital, Central South University, Hunan, 410013, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Junhui Li
- Transplantation Center, The Third Xiangya Hospital, Central South University, Hunan, 410013, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Min Yang
- Transplantation Center, The Third Xiangya Hospital, Central South University, Hunan, 410013, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Ke Cheng
- Transplantation Center, The Third Xiangya Hospital, Central South University, Hunan, 410013, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Yingzi Ming
- Transplantation Center, The Third Xiangya Hospital, Central South University, Hunan, 410013, Changsha, China.
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China.
| |
Collapse
|
46
|
Vanderborght B, De Muynck K, Gijbels E, Lefere S, Scott CL, Guilliams M, Beschin A, Vinken M, Verhelst X, Geerts A, Van Vlierberghe H, Devisscher L. Transient Kupffer cell depletion and subsequent replacement by infiltrating monocyte-derived cells does not alter the induction or progression of hepatocellular carcinoma. Int J Cancer 2023; 152:2615-2628. [PMID: 36912275 DOI: 10.1002/ijc.34505] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 01/17/2023] [Accepted: 02/09/2023] [Indexed: 03/14/2023]
Abstract
Due to a combination of rapid disease progression and the lack of curative treatment options, hepatocellular carcinoma (HCC) is one of the deadliest cancers worldwide. Infiltrated, monocyte-derived, tumor-associated macrophages are known to play a role in HCC pathogenesis, but the involvement of Kupffer cells (KCs) remains elusive. Here, we used the Clec4F-diphteria toxin receptor transgenic mouse model to specifically investigate the effect of KC depletion on HCC initiation, progression and neoplastic growth following liver resection. For this purpose, several HCC mouse models with varying underlying etiologies were used and partial hepatectomy was performed. Our results show that in HCC, developed on a fibrotic or non-alcoholic steatohepatitis background, depletion of embryonic KCs at the onset of HCC induction and the subsequent replacement by monocyte-derived KCs does not affect the tumor burden, tumor microenvironment or the phenotype of isolated KCs at end-stage disease. In non-chronic liver disease-associated diethylnitrosamine-induced HCC, ablation of Clec4F+ KCs did not alter tumor progression or neoplastic growth following liver resection. Our results show that temporal ablation of resident KCs does not impact HCC pathogenesis, neither in the induction phase nor in advanced disease, and indicate that bone marrow-derived KCs are able to swiftly repopulate the available KC niche and adopt their phenotype.
Collapse
Affiliation(s)
- Bart Vanderborght
- Hepatology Research Unit, Department of Internal Medicine and Pediatrics, Liver Research Center Ghent, Faculty of Medicine and Health Sciences, Ghent University, Ghent, 9000, Belgium
- Gut-Liver Immunopharmacology Unit, Department of Basic and Applied Medical Sciences, Liver Research Center Ghent, Faculty of Medicine and Health Sciences, Ghent University, Ghent, 9000, Belgium
| | - Kevin De Muynck
- Hepatology Research Unit, Department of Internal Medicine and Pediatrics, Liver Research Center Ghent, Faculty of Medicine and Health Sciences, Ghent University, Ghent, 9000, Belgium
- Gut-Liver Immunopharmacology Unit, Department of Basic and Applied Medical Sciences, Liver Research Center Ghent, Faculty of Medicine and Health Sciences, Ghent University, Ghent, 9000, Belgium
| | - Eva Gijbels
- Gut-Liver Immunopharmacology Unit, Department of Basic and Applied Medical Sciences, Liver Research Center Ghent, Faculty of Medicine and Health Sciences, Ghent University, Ghent, 9000, Belgium
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, 1090, Belgium
| | - Sander Lefere
- Hepatology Research Unit, Department of Internal Medicine and Pediatrics, Liver Research Center Ghent, Faculty of Medicine and Health Sciences, Ghent University, Ghent, 9000, Belgium
| | - Charlotte L Scott
- Department of Biomedical Molecular Biology, Faculty of Science, Ghent University, Ghent, 9000, Belgium
- Laboratory of Myeloid Cell Biology in Tissue Damage and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, 9000, Belgium
| | - Martin Guilliams
- Department of Biomedical Molecular Biology, Faculty of Science, Ghent University, Ghent, 9000, Belgium
- Laboratory of Myeloid Cell Biology in Tissue Homeostasis and Regeneration, VIB-UGent Center for Inflammation Research, Ghent, 9000, Belgium
| | - Alain Beschin
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, 1090, Belgium
- Myeloid Cell Immunology Laboratory, Vrije Universiteit Brussel, Brussels, 1090, Belgium
| | - Mathieu Vinken
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, 1090, Belgium
| | - Xavier Verhelst
- Hepatology Research Unit, Department of Internal Medicine and Pediatrics, Liver Research Center Ghent, Faculty of Medicine and Health Sciences, Ghent University, Ghent, 9000, Belgium
- Department of Gastroenterology and Hepatology, Ghent University Hospital, Ghent, 9000, Belgium
| | - Anja Geerts
- Hepatology Research Unit, Department of Internal Medicine and Pediatrics, Liver Research Center Ghent, Faculty of Medicine and Health Sciences, Ghent University, Ghent, 9000, Belgium
- Department of Gastroenterology and Hepatology, Ghent University Hospital, Ghent, 9000, Belgium
| | - Hans Van Vlierberghe
- Hepatology Research Unit, Department of Internal Medicine and Pediatrics, Liver Research Center Ghent, Faculty of Medicine and Health Sciences, Ghent University, Ghent, 9000, Belgium
- Department of Gastroenterology and Hepatology, Ghent University Hospital, Ghent, 9000, Belgium
| | - Lindsey Devisscher
- Gut-Liver Immunopharmacology Unit, Department of Basic and Applied Medical Sciences, Liver Research Center Ghent, Faculty of Medicine and Health Sciences, Ghent University, Ghent, 9000, Belgium
| |
Collapse
|
47
|
Alabdulaali B, Al-rashed F, Al-Onaizi M, Kandari A, Razafiarison J, Tonui D, Williams MR, Blériot C, Ahmad R, Alzaid F. Macrophages and the development and progression of non-alcoholic fatty liver disease. Front Immunol 2023; 14:1195699. [PMID: 37377968 PMCID: PMC10291618 DOI: 10.3389/fimmu.2023.1195699] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
The liver is the site of first pass metabolism, detoxifying and metabolizing blood arriving from the hepatic portal vein and hepatic artery. It is made up of multiple cell types, including macrophages. These are either bona fide tissue-resident Kupffer cells (KC) of embryonic origin, or differentiated from circulating monocytes. KCs are the primary immune cells populating the liver under steady state. Liver macrophages interact with hepatocytes, hepatic stellate cells, and liver sinusoidal endothelial cells to maintain homeostasis, however they are also key contributors to disease progression. Generally tolerogenic, they physiologically phagocytose foreign particles and debris from portal circulation and participate in red blood cell clearance. However as immune cells, they retain the capacity to raise an alarm to recruit other immune cells. Their aberrant function leads to the development of non-alcoholic fatty liver disease (NAFLD). NAFLD refers to a spectrum of conditions ranging from benign steatosis of the liver to steatohepatitis and cirrhosis. In NAFLD, the multiple hit hypothesis proposes that simultaneous influences from the gut and adipose tissue (AT) generate hepatic fat deposition and that inflammation plays a key role in disease progression. KCs initiate the inflammatory response as resident immune effectors, they signal to neighbouring cells and recruit monocytes that differentiated into recruited macrophages in situ. Recruited macrophages are central to amplifying the inflammatory response and causing progression of NAFLD to its fibro-inflammatory stages. Given their phagocytic capacity and their being instrumental in maintaining tissue homeostasis, KCs and recruited macrophages are fast-becoming target cell types for therapeutic intervention. We review the literature in the field on the roles of these cells in the development and progression of NAFLD, the characteristics of patients with NAFLD, animal models used in research, as well as the emerging questions. These include the gut-liver-brain axis, which when disrupted can contribute to decline in function, and a discussion on therapeutic strategies that act on the macrophage-inflammatory axis.
Collapse
Affiliation(s)
- Bader Alabdulaali
- Dasman Diabetes Institute, Kuwait City, Kuwait
- Ministry of Health, Kuwait City, Kuwait
| | | | - Mohammed Al-Onaizi
- Dasman Diabetes Institute, Kuwait City, Kuwait
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Anwar Kandari
- Dasman Diabetes Institute, Kuwait City, Kuwait
- Ministry of Health, Kuwait City, Kuwait
| | - Joanna Razafiarison
- INSERM UMR-S1151, CNRS UMR-S8253, Université Paris Cité, Institut Necker Enfants Malades, Paris, France
| | - Dorothy Tonui
- INSERM UMR-S1151, CNRS UMR-S8253, Université Paris Cité, Institut Necker Enfants Malades, Paris, France
| | | | - Camille Blériot
- INSERM UMR-S1151, CNRS UMR-S8253, Université Paris Cité, Institut Necker Enfants Malades, Paris, France
- Inserm U1015, Gustave Roussy, Villejuif, France
| | | | - Fawaz Alzaid
- Dasman Diabetes Institute, Kuwait City, Kuwait
- INSERM UMR-S1151, CNRS UMR-S8253, Université Paris Cité, Institut Necker Enfants Malades, Paris, France
| |
Collapse
|
48
|
Goto Y, Mizobuchi H. Pathological roles of macrophages in Leishmania infections. Parasitol Int 2023; 94:102738. [PMID: 36738983 DOI: 10.1016/j.parint.2023.102738] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 01/31/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
Macrophages are the major host cells for Leishmania parasites, and determine the fate of infection by either limiting or allowing growth of the parasites, resulting in development or control of leishmaniasis, respectively. They also play important roles in causing pathological outcomes during Leishmania infection. The pathophysiology is complex and include a wide variety of molecular and cellular responses including enhancement of inflammatory responses by releasing cytokines, causing damages to surrounding cells by reactive oxygen species, or disordered phagocytosis of other cells. It is of note that disease severity in leishmaniasis sometimes does not correlate with parasite burdens, indicating that pathological roles of macrophages are not necessarily linked to their parasite-killing activities that are often defined by M1/M2 status. Here, we review the roles of macrophages in leishmaniasis with a focus on their pathological mechanisms in disease development.
Collapse
Affiliation(s)
- Yasuyuki Goto
- Laboratory of Molecular Immunology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan.
| | - Haruka Mizobuchi
- Laboratory of Molecular Immunology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| |
Collapse
|
49
|
Feng H, Dai W. Case Report: FDG-PET/CT findings in co-infection of visceral leishmaniasis and chronic hepatitis B. Front Cell Infect Microbiol 2023; 13:1175897. [PMID: 37325515 PMCID: PMC10264663 DOI: 10.3389/fcimb.2023.1175897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/16/2023] [Indexed: 06/17/2023] Open
Abstract
Visceral leishmaniasis is an opportunistic infection in immunocompromised patients. Herein, we report a case of an adult male patient with a persistent fever of unknown origin, along with chronic hepatitis B. The patient underwent bone marrow aspiration twice, which revealed hemophagocytosis. Abdomen enhanced CT revealed splenomegaly with a persistent strengthening of multiple nodules, and hemangiomas were diagnosed. A subsequent 18-fluoro-deoxyglucose (18F-FDG) PET/CT scan, which was implemented to search for the reason for the fever, showed diffuse splenic disease uptake, and splenic lymphoma was considered as the diagnosis. His clinical symptoms improved after receiving hemophagocytic lymphohistiocytosis (HLH) chemotherapy. However, the patient was readmitted for fever again only 2 months later. Splenectomy surgery is performed to confirm the diagnosis and classification of lymphoma. Visceral leishmaniasis was eventually diagnosed in a spleen specimen and the third bone marrow biopsy. He received treatment with lipid amphotericin B and remained recurrence-free for 1 year. In this paper, we aim to provide detailed information that will help further our understanding of the clinical symptoms and radiographic findings of visceral leishmaniasis.
Collapse
Affiliation(s)
| | - Wenli Dai
- Department of Nuclear Medicine, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, China
| |
Collapse
|
50
|
Zhang X, Ohayon-Steckel L, Coppin E, Johny E, Dasari A, Florentin J, Vasamsetti S, Dutta P. Epidermal Growth Factor Receptor in Hepatic Endothelial Cells Suppresses MCP-1-Dependent Monocyte Recruitment in Diabetes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1363-1371. [PMID: 36946774 PMCID: PMC10121888 DOI: 10.4049/jimmunol.2200557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 02/27/2023] [Indexed: 03/23/2023]
Abstract
Insulin resistance is a compromised response to insulin in target tissues such as liver. Emerging evidence shows that vascular endothelial cells (ECs) are critical in mediating glucose metabolism. However, how liver ECs can regulate inflammation in the setting of insulin resistance is still unknown. Using genome-wide transcriptome analysis of ECs isolated from diabetic mice, we found enrichment of the genes involved in epidermal growth factor receptor (Egfr) signaling. In line with this, hepatic sinusoidal ECs in diabetic mice had elevated levels of Egfr expression. Interestingly, we found an increased number of hepatic myeloid cells, especially macrophages, and systemic glucose intolerance in Cdh5Cre/+Egfrfl/fl mice lacking Egfr in ECs compared with littermate control mice with type II diabetes. Egfr deficiency upregulated the expression of MCP-1 in hepatic sinusoidal ECs. This resulted in augmented monocyte recruitment and macrophage differentiation in Cdh5Cre/+Egfrfl/fl mice compared with littermate control mice as determined by a mouse model of parabiosis. Finally, MCP-1 neutralization and hepatic macrophage depletion in Cdh5Cre/+Egfrfl/fl mice resulted in a reduced number of hepatic macrophages and ameliorated glucose intolerance compared with the control groups. Collectively, these results demonstrate a protective endothelial Egfr signaling in reducing monocyte-mediated hepatic inflammation and glucose intolerance in type II diabetic mice.
Collapse
Affiliation(s)
- Xinyi Zhang
- Department of Cardiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Lee Ohayon-Steckel
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Emilie Coppin
- Regeneration in Hematopoiesis, Institute for Immunology, TU Dresden, Dresden, Germany
- Immunology of Aging, Leibniz Institute on Aging – Fritz Lipmann Institute, Jena, Germany
| | - Ebin Johny
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Ankush Dasari
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jonathan Florentin
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Sathish Vasamsetti
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Partha Dutta
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Pittsburgh VA Medical Center-University Drive, University Drive C, Pittsburgh, PA, 15213
| |
Collapse
|