1
|
Sun Y, Guo Z, Huo Y, Zhang H, Li T, Wang P, Han W. CMTM7 inhibits TLR4 signaling pathway via promoting Rab5 activation and alleviates acute liver injury. Cell Mol Life Sci 2025; 82:229. [PMID: 40490565 DOI: 10.1007/s00018-025-05748-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 03/26/2025] [Accepted: 05/09/2025] [Indexed: 06/11/2025]
Abstract
The activation of macrophages mediated by TLR4 is crucial for innate immune responses, while the regulatory mechanisms of TLR4 are still under investigation. This study demonstrates that CMTM7 inhibits TLR4 pathway activation in macrophages and exerts a protective role in acute liver injury (ALI). CMTM7 is highly expressed in monocytes/macrophages, which is downregulated upon LPS stimulation. CMTM7 inhibits LPS/HMGB1-induced activation of the TLR4 pathway in macrophages. Mechanistically, CMTM7 promotes the binding between Rab5 and Gapex5, leading to the generation of GTP-Rab5, which facilitates the internalization and degradation of TLR4, thereby inhibiting TLR4 signaling activation. Utilizing Cmtm7 myeloid conditional knockout mice, we confirmed the protective role of CMTM7 in ALI and highlighted its therapeutic potential through the adoptive transfer of CMTM7-overexpressing macrophages. This study elucidates a novel regulatory mechanism of TLR4 signaling transduction and provides a novel therapeutic strategy for ALI treatment.
Collapse
Affiliation(s)
- Yingzhe Sun
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University Health Science Center, Beijing, China
- Peking University Center for Human Disease Genomics, Beijing, China
- Key Laboratory of Geriatrics (Hepatobiliary Diseases), China General Technology Group, Beijing Electric Power Hospital, State Grid Corporation of China, Capital Medical University, Beijing, China
| | - Zixia Guo
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University Health Science Center, Beijing, China
- Peking University Center for Human Disease Genomics, Beijing, China
| | - Yangbo Huo
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University Health Science Center, Beijing, China
- Peking University Center for Human Disease Genomics, Beijing, China
| | - Hanxiao Zhang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University Health Science Center, Beijing, China
- Peking University Center for Human Disease Genomics, Beijing, China
| | - Ting Li
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University Health Science Center, Beijing, China
- Peking University Center for Human Disease Genomics, Beijing, China
| | - Pingzhang Wang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University Health Science Center, Beijing, China
- Peking University Center for Human Disease Genomics, Beijing, China
- Key Laboratory of Geriatrics (Hepatobiliary Diseases), China General Technology Group, Beijing Electric Power Hospital, State Grid Corporation of China, Capital Medical University, Beijing, China
| | - Wenling Han
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University Health Science Center, Beijing, China.
- Peking University Center for Human Disease Genomics, Beijing, China.
- Key Laboratory of Geriatrics (Hepatobiliary Diseases), China General Technology Group, Beijing Electric Power Hospital, State Grid Corporation of China, Capital Medical University, Beijing, China.
| |
Collapse
|
2
|
Poole LG, Wei Z, Schulte A, Cline HM, Bernard MP, Buchweitz JP, McGill MR, Luyendyk JP. Kupffer cell expression of macrophage receptor with collagenous structure modulates macrophage gene induction and limits acute liver injury. Toxicol Sci 2025; 205:417-427. [PMID: 40117216 PMCID: PMC12118956 DOI: 10.1093/toxsci/kfaf037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2025] Open
Abstract
Macrophages displaying a pro-repair and anti-inflammatory polarization have been implicated in resolution of acute liver injury. Macrophage receptor with collagenous structure (MARCO) expression marks tolerogenic hepatic macrophages and is expressed by pro-resolution macrophages in the injured liver. We tested the hypothesis that MARCO promotes repair of the acetaminophen (APAP)-injured liver. Robust and sustained induction of MARCO mRNA and protein expression was evident in livers of mice challenged with a hepatotoxic dose of APAP (i.e. 300 mg/kg), whereas hepatic MARCO induction failed in mice with APAP-induced liver failure (i.e. 600 mg/kg). Serum proteomics identified a significant increase in serum MARCO levels in surviving acute liver failure (ALF) patients, but not in ALF patients who died. MARCO expression was high in F480+ liver macrophages, and MARCO deficiency reduced macrophage expression of pro-resolution markers such as Gpnmb and Mertk during the repair phase (i.e. 48 h). The results suggested a delay in necrosis resolution along with a trend toward increased mortality in APAP-challenged MARCO-/- mice. Notably, a robust increase in peak hepatic injury (i.e. 6- to 24-h post-APAP challenge) was evident in MARCO-/- mice, which could not be ascribed to differences in NAPQI/APAP-adduct generation nor changes in hepatic neutrophil/macrophage numbers. Interestingly, a reduction in hepatic CD11c+ cells, shown previously to limit APAP-induced liver injury, was evident 24 h after APAP challenge in MARCO-/- mice. The results indicate that MARCO deficiency worsens APAP-induced acute liver injury in mice and provide experimental and initial translational evidence linking MARCO induction to positive outcomes in acute liver injury.
Collapse
Affiliation(s)
- Lauren G Poole
- Department of Pathobiology & Diagnostic Investigation, Michigan State University, East Lansing, MI 48824, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, United States
| | - Zimu Wei
- Department of Pathobiology & Diagnostic Investigation, Michigan State University, East Lansing, MI 48824, United States
| | - Anthony Schulte
- Department of Pathobiology & Diagnostic Investigation, Michigan State University, East Lansing, MI 48824, United States
| | - Holly M Cline
- Department of Pathobiology & Diagnostic Investigation, Michigan State University, East Lansing, MI 48824, United States
| | - Matthew P Bernard
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI 48824, United States
| | - John P Buchweitz
- Department of Pathobiology & Diagnostic Investigation, Michigan State University, East Lansing, MI 48824, United States
| | - Mitchell R McGill
- Department of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - James P Luyendyk
- Department of Pathobiology & Diagnostic Investigation, Michigan State University, East Lansing, MI 48824, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, United States
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI 48824, United States
| |
Collapse
|
3
|
Xu F, Jansakun C, Li G, Biswas U, Poschet G, Staffer S, Tuma-Kellner S, Nakchbandi I, Merle U, Chamulitrat W. Myeloid-specific deficiency of group VIA calcium-independent phospholipase A2 preconditions myeloid cells for injury resolution after acetaminophen exposure. Biomed Pharmacother 2025; 187:118146. [PMID: 40344700 DOI: 10.1016/j.biopha.2025.118146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 05/02/2025] [Accepted: 05/05/2025] [Indexed: 05/11/2025] Open
Abstract
Genetic PLA2G6 variants are associated with C-reactive protein in humans. Myeloid-specific PLA2G6-deficient (Pla2g6M-/-) mice show increased hepatic myeloperoxidase and recruitment of granulocytes in response to lipopolysaccharide (LPS). We hypothesized that Pla2g6M-/- mice could be protected from acetaminophen (APAP) hepatotoxicity whereby neutrophils, eosinophils, and alternatively activated macrophages are reportedly protective. Herein, Pla2g6M-/- mice treated with 300 mg/kg APAP for 24 h showed attenuated hepatic necrosis and plasma cytokines, and with elevated levels of Ly6Clo in peripheral blood mononuclear cells and plasma lipoxin A4. Remarkably, bone-marrow-derived macrophages (BMDMs) from untreated Pla2g6M-/- mice exhibited elevated baseline expression of cPLA2α, NOX2, Rac1, Arg-1, phospho-MLKL, and iNOS protein, which was exacerbated by LPS in vitro. APAP administration preconditioned Pla2g6M-/- BMDMs for further activation of enzymes involving in phagocytosis (Rac1 and phospho-MLKL) and eicosanoids (COX2 and A15LOXB). Pla2g6M-/- BMDMs showed an increased release of pro-resolution lipid mediators lipoxin A4, PGE2, and 15d-PGJ2, which was further elevated by LPS in vitro or APAP in vivo. Phagocytic gene signatures (myeloperoxidase and NOX2) were also upregulated in livers of untreated and APAP-treated Pla2g6M-/- mice. APAP protection in Pla2g6M-/- mice was associated with increased proportion of neutrophils (Ly6G), eosinophils (eosinophilic cationic protein), and M2 macrophages (CD206) in/at the portal tract and central vein as determined by immunohistochemistry. Thus, myeloid-specific PLA2G6 deficiency preconditioned macrophages for eicosanoid and phagocytic pathways rendering protection against APAP hepatotoxicity. Our results may be applicable to patients with PLA2G6 mutations, and PLA2G6 inhibition specifically in myeloid cells may represent a new strategy to alleviate APAP poisoning.
Collapse
Affiliation(s)
- Feng Xu
- Internal Medicine IV, Heidelberg University Hospital, Im Neuenheimer Feld 410, Heidelberg 69120, Germany; Gastrointestinal Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, PR China
| | - Chutima Jansakun
- Internal Medicine IV, Heidelberg University Hospital, Im Neuenheimer Feld 410, Heidelberg 69120, Germany; School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80161, Thailand
| | - Gang Li
- Internal Medicine IV, Heidelberg University Hospital, Im Neuenheimer Feld 410, Heidelberg 69120, Germany; Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Uddipta Biswas
- Internal Medicine IV, Heidelberg University Hospital, Im Neuenheimer Feld 410, Heidelberg 69120, Germany
| | - Gernot Poschet
- Metabolomics Core Technology Platform, Centre for Organismal Studies, University of Heidelberg, Heidelberg 69120, Germany
| | - Simone Staffer
- Internal Medicine IV, Heidelberg University Hospital, Im Neuenheimer Feld 410, Heidelberg 69120, Germany
| | - Sabine Tuma-Kellner
- Internal Medicine IV, Heidelberg University Hospital, Im Neuenheimer Feld 410, Heidelberg 69120, Germany
| | - Inaam Nakchbandi
- Max-Planck Institute of Biochemistry and University of Heidelberg, Im Neuenheimer Feld 305, Heidelberg 69120, Germany
| | - Uta Merle
- Internal Medicine IV, Heidelberg University Hospital, Im Neuenheimer Feld 410, Heidelberg 69120, Germany
| | - Walee Chamulitrat
- Internal Medicine IV, Heidelberg University Hospital, Im Neuenheimer Feld 410, Heidelberg 69120, Germany.
| |
Collapse
|
4
|
Zhao X, Li Y, Yang L, Chen X, Zhang J, Chen T, Wang H, Li F, Cheng C, Wu J, Cong J, Yin W, Li J, Wang X. GPR35 prevents drug-induced liver injury via the Gαs-cAMP-PKA axis in macrophages. Cell Mol Life Sci 2025; 82:219. [PMID: 40437264 PMCID: PMC12119454 DOI: 10.1007/s00018-025-05751-4] [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: 12/10/2024] [Revised: 03/31/2025] [Accepted: 05/13/2025] [Indexed: 06/01/2025]
Abstract
Acetaminophen (APAP) overdose induces acute liver injury and represents the most frequent cause of drug-induced liver injury worldwide. Macrophage-mediated inflammation plays detrimental roles during the early stage of liver injury. However, the potential targets regulating inflammation to improve drug-induced liver injury remains undefined. In this study, we reported that G protein-coupled receptor 35 (GPR35) improves drug-induced liver injury by blocking macrophage-mediated inflammation via the Gαs-cyclic AMP-protein kinase A (Gαs-cAMP-PKA) pathway. The ablation of GPR35 exacerbates APAP-induced liver injury, characterized by higher levels of alanine aminotransferase and aspartate aminotransferase in sera, larger damaged areas, and increased levels of pro-inflammatory cytokines. More hepatic macrophages appeared in the inflamed liver of mice with GPR35 deficiency. In contrast, the agonists of GPR35 alleviated APAP-induced liver injury. The depletion of macrophages abolished GPR35-mediated protection. Mechanistically, GPR35 ablation facilitated the activation of pro-inflammatory AKT, MAPK, and NF-κB signaling pathways at the downstream of Toll-like receptors in macrophages. GPR35 agonists activated Gαs-cAMP-PKA signaling to inhibit the activation of these pro-inflammatory signaling pathways and then suppress the inflammatory response in macrophages. Thus, our findings demonstrate that GPR35 prevents drug-induced liver injury by blocking macrophage-mediated inflammation via the Gαs-cAMP-PKA pathway, indicating that GPR35 is a potential target for the development of novel medicines that control drug-induced liver injury.
Collapse
Affiliation(s)
- Xueqin Zhao
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, #81 Meishan Road, Hefei, 230032, Anhui, China
| | - Yuanhao Li
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, #81 Meishan Road, Hefei, 230032, Anhui, China
| | - Liu Yang
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, #81 Meishan Road, Hefei, 230032, Anhui, China
| | - Xi Chen
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, #81 Meishan Road, Hefei, 230032, Anhui, China
| | - Jialong Zhang
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, #81 Meishan Road, Hefei, 230032, Anhui, China
| | - Tong Chen
- School of Life Sciences, Anhui Medical University, #81 Meishan Road, Hefei, 230032, Anhui, China
| | - Haoqi Wang
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, #81 Meishan Road, Hefei, 230032, Anhui, China
| | - Fei Li
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, #81 Meishan Road, Hefei, 230032, Anhui, China
| | - Chen Cheng
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, #81 Meishan Road, Hefei, 230032, Anhui, China
| | - Jingjing Wu
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Jingjing Cong
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, #81 Meishan Road, Hefei, 230032, Anhui, China
| | - Wenwei Yin
- Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China.
| | - Jing Li
- School of Life Sciences, Anhui Medical University, #81 Meishan Road, Hefei, 230032, Anhui, China.
| | - Xuefu Wang
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, #81 Meishan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
5
|
Chen L, Guillot A, Tacke F. Reviewing the function of macrophages in liver disease. Expert Rev Gastroenterol Hepatol 2025:1-17. [PMID: 40387555 DOI: 10.1080/17474124.2025.2508963] [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: 12/03/2024] [Revised: 04/10/2025] [Accepted: 05/08/2025] [Indexed: 05/20/2025]
Abstract
INTRODUCTION The liver is a central metabolic organ, but is also hosting a unique immune microenvironment to sustain homeostasis and proper defense measures against injury threats in healthy individuals. Liver macrophages, mostly represented by the tissue-resident Kupffer cells and bone marrow- or monocyte-derived macrophages, are intricately involved in various aspects of liver homeostasis and disease, including tissue injury, inflammation, fibrogenesis and repair mechanisms. AREAS COVERED We review recent findings on defining the liver macrophage landscape and their functions in liver diseases with the aim of highlighting potential targets for therapeutic interventions. A comprehensive literature search in PubMed and Google Scholar was conducted to identify relevant literature up to date. EXPERT OPINION Liver macrophages orchestrate key homeostatic and pathogenic processes in the liver. Thus, targeting liver macrophages represents an attractive strategy for drug development, e.g. to ameliorate liver inflammation, steatohepatitis or fibrosis. However, translation from fundamental research to therapies remains challenging due to the versatile nature of the liver macrophage compartment. Recent and major technical advances such as single-cell and spatially-resolved omics approaches deepened our understanding of macrophage biology at a molecular level. Yet, further studies are needed to identify suitable, etiology- and stage-dependent strategies for the treatment of liver diseases.
Collapse
Affiliation(s)
- Lanlan Chen
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Adrien Guillot
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
6
|
Li R, Shu Y, Yan Y, Zhu J, Cheng Z, Zhang J, Zhu L, Qiao Y, Sun Q. Caveolin-1 Deficiency in Macrophages Alleviates Carbon Tetra-Chloride-Induced Acute Liver Injury in Mice. Int J Mol Sci 2025; 26:4903. [PMID: 40430042 PMCID: PMC12112502 DOI: 10.3390/ijms26104903] [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: 03/28/2025] [Revised: 05/08/2025] [Accepted: 05/11/2025] [Indexed: 05/29/2025] Open
Abstract
Bone marrow-derived macrophages (BMMs) exhibit dynamic behavior and functional capabilities in response to specific microenvironmental stimuli. Recent investigations have proved that BMMs play crucial roles in promoting necrotic lesion resolution. Despite substantial advancements in understanding their activation and interaction with injured livers, researchers face challenges to develop effective treatments based on manipulating BMMs function. Caveolin-1 (Cav-1) is the major structural protein on the plasma membrane. We previously reported that Cav-1 knockout (KO) mice exhibited less functional damage and necrosis in carbon tetrachloride (CCl4)-induced liver injury. We hypothesize that the activation and recruitment of BMMs are involved in the resolution of necrotic lesions in Cav-1 KO mice. Wild-type (WT) and Cav-1 KO mice were injected with CCl4 (10% v/v) to induce acute liver injury model. Blood samples and hepatic tissues were harvested for serum alanine transaminase (ALT) activity assessment, histopathological examination through hematoxylin-eosin (H&E) staining, and BMMs subpopulation analysis via flow cytometry. Then, primary BMMs were isolated and cultured to investigate the effect of Cav-1 on BMMs polarization, migration, and activation of STAT3 signal pathway. Validation of hepatic macrophage depletion was induced by administrating clodronate liposomes (CLs), and BMMs reconstitution was evaluated by EGFP labelled BMMs. Following this, hepatic macrophages were depleted by CLs, BMMs were isolated from Cav-1 KO, and WT mice were cultured and administrated to evaluate the protective role of Cav-1-deleted BMMs on the resolution of hepatocellular necrosis and apoptosis in acute liver injury. The BMMs ratio significantly increased from 2.12% (1D), 4.38% (1W), and 5.38% (2W) in oil control mice to 7.17%, 14.90%, and 19.30% in CCl4-treated mice (p < 0.01 or p < 0.001). Concurrently, Cav-1 positive BMMs exhibited a marked elevation from 6.41% at 1D to 24.90% by 2W (p = 0.0228). Cav-1 KO exerted protective effects by reducing serum ALT by 26% (p = 0.0265) and necrotic areas by 28% (p = 0.0220) and enhancing BMMs infiltration by 60% (p = 0.0059). In vitro, Cav-1 KO BMMs showed a decrease in CD206 fluorescence intensity (p < 0.001), a time-dependent upregulation of arginase-1 mRNA (p < 0.05 or p < 0.01), a 1.22-fold increase in phosphorylated STAT3 (p = 0.0036), and impaired wound healing from 12 to 24 h (p < 0.001). The macrophage-depleting action in livers by CL injection persists for a minimum of 48 h. Administrated EGFP+ BMMs emerged as the predominant population following CL injection for a duration of 48 h. Following clodronate liposome-mediated hepatic macrophage depletion, the adoptive transfer of Cav-1 KO BMMs demonstrated therapeutic efficacy in CCl4-induced acute liver injury. In CCl4-induced acute liver injury, the adoptive transfer of Cav-1 KO BMMs reduced necrosis by 12.8% (p = 0.0105), apoptosis by 25.2% (p = 0.0127), doubled macrophages infiltration (p = 0.0269), and suppressed CXCL9/10 mRNA expression (p = 0.0044 or p = 0.0385). BMMs play a key role in the resolution of liver necrotic lesions in CCl4-induced acute liver injury. Cav-1 depletion attenuates hepatocellular necrosis and apoptosis by accelerating BMMs recruitment and M2 polarization. Cav-1 in macrophages may represent a potential therapeutic target for acute liver injury.
Collapse
Affiliation(s)
- Ruirui Li
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (R.L.); (Y.S.); (Y.Y.); (J.Z.); (Z.C.)
| | - Yixue Shu
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (R.L.); (Y.S.); (Y.Y.); (J.Z.); (Z.C.)
| | - Yulin Yan
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (R.L.); (Y.S.); (Y.Y.); (J.Z.); (Z.C.)
| | - Junyi Zhu
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (R.L.); (Y.S.); (Y.Y.); (J.Z.); (Z.C.)
| | - Zilu Cheng
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (R.L.); (Y.S.); (Y.Y.); (J.Z.); (Z.C.)
| | - Jie Zhang
- Laboratory Animal Center, Capital Medical University, Beijing 100069, China; (J.Z.); (L.Z.); (Y.Q.)
| | - Liming Zhu
- Laboratory Animal Center, Capital Medical University, Beijing 100069, China; (J.Z.); (L.Z.); (Y.Q.)
| | - Yanhua Qiao
- Laboratory Animal Center, Capital Medical University, Beijing 100069, China; (J.Z.); (L.Z.); (Y.Q.)
| | - Quan Sun
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (R.L.); (Y.S.); (Y.Y.); (J.Z.); (Z.C.)
- Laboratory Animal Center, Capital Medical University, Beijing 100069, China; (J.Z.); (L.Z.); (Y.Q.)
| |
Collapse
|
7
|
Kang Z, Xie R, Cui Y, Chen Z, Li J, Lv J, Ye W, Zhao P, Zhang K, Hong J, Qu H. Macrophage PKM2 depletion ameliorates hepatic inflammation and acute liver injury in mice. Front Pharmacol 2025; 16:1546045. [PMID: 40351417 PMCID: PMC12062095 DOI: 10.3389/fphar.2025.1546045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 03/31/2025] [Indexed: 05/14/2025] Open
Abstract
Introduction Pyruvate kinase M2 (PKM2), the rate-limiting enzyme of glycolysis, plays a critical role in macrophage activation and a broad spectrum of chronic liver diseases. However, whether PKM2 contributes to the pathogenesis of acute liver injury (ALI) remains largely unexplored. Methods PKM2 expression was assessed in human and mouse ALI livers. Macrophage-specific PKM2 knockout mice were challenged by two independent ALI models, induced by acetaminophen (APAP) and lipopolysaccharide/D-galactosamine (LPS/D-GalN), to explore the role and regulatory mechanism of macrophage PKM2 in ALI progression. Results By bioinformatic screening and analysis of ALI liver, we found that PKM2 was significantly upregulated in the liver tissues of ALI patients and mice. Immunofluorescence staining further demonstrated that PKM2 was markedly upregulated in macrophages during ALI progression. Notably, macrophage PKM2 depletion effectively alleviated APAP- and LPS/D-GalN-induced ALI, as demonstrated by ameliorated immune cells infiltration, pro-inflammatory mediators, and hepatocellular cell death. PKM2-deficient macrophages showed M2 anti-inflammatory polarization in vivo and in vitro. Furthermore, PKM2 deletion limited HIF-1α signaling and aerobic glycolysis of macrophages, which thereby attenuated macrophage pro-inflammatory activation and hepatocyte injury. Pharmacological PKM2 antagonist efficiently ameliorated liver injury and prolonged the survival of mice in APAP-induced ALI model. Discussion Our study highlights the pivotal role of macrophage PKM2 in advancing ALI, and therapeutic targeting of PKM2 may serve as a novel strategy to combat ALI.
Collapse
Affiliation(s)
- Ziwei Kang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, China
| | - Ruoyan Xie
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, China
| | - Yiming Cui
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, China
| | - Zhiwei Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Jincheng Li
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jinyu Lv
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, China
| | - Weijia Ye
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Peixin Zhao
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, China
| | - Keke Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, China
| | - Jian Hong
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, China
| | - Hengdong Qu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
8
|
Berasain C. New Insights and Open Questions on the Molecular and Cellular Crosstalk Governing Hepatocyte Proliferation. Cell Mol Gastroenterol Hepatol 2025:101509. [PMID: 40245923 DOI: 10.1016/j.jcmgh.2025.101509] [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: 03/16/2025] [Accepted: 03/20/2025] [Indexed: 04/19/2025]
Affiliation(s)
- Carmen Berasain
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, Carlos III Health Institute), Madrid, Spain.
| |
Collapse
|
9
|
Fan M, Xu Y, Wu B, Long J, Liu C, Liang Z, Zhang R, Liu Z, Wang C. Geniposidic Acid Targeting FXR "S332 and H447" Mediated Conformational Change to Upregulate CYPs and miR-19a-3p to Ameliorate Drug-Induced Liver Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409107. [PMID: 39998442 PMCID: PMC12005789 DOI: 10.1002/advs.202409107] [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: 08/04/2024] [Revised: 01/03/2025] [Indexed: 02/26/2025]
Abstract
Drug-induced liver injury (DILI), caused by chemical drugs and traditional Chinese medicine, often leads to severe outcomes like liver failure due to a lack of early detection markers. Farnesoid X receptor (FXR), a key regulator of bile acid (BA) and cholesterol metabolism, is a potential therapeutic target. This study investigates the pathogenesis, markers, and treatment strategies for DILI, focusing on the hepatoprotective effects of geniposidic acid (GPA) from Gardenia jasminoides J. Ellis. Using cellular and animal models of acute and chronic DILI induced by acetaminophen and triptolide, we explored GPA's mechanisms in BA and cholesterol metabolism. Lipidomic and BA analyses revealed that GPA alleviates DILI by enhancing bile acid synthesis and transport via FXR activation. Experiments using AAV-shFXR, Fxr- / - mice and molecular assays demonstrated that GPA targets Ser332 and His447 on FXR ligand-binding domain, promoting FXR nuclear translocation and initiating cytochrome P450 proteins (CYPs) transcriptional activation for BA metabolism. Additionally, miRNA sequencing and RNA-pulldown assays showed that GPA-activated FXR upregulates miR-19a-3p, binding to LXR 3'UTR to inhibit cholesterol production. These findings reveal the GPA-FXR "structure-target" relationship, highlighting a dual mechanism in which GPA promotes CYPs-mediated bile acid metabolism and miR-19a-3p-mediated cholesterol synthesis inhibition, providing a basis for FXR-targeted DILI therapies.
Collapse
Affiliation(s)
- Minqi Fan
- State Key Laboratory of Traditional Chinese Medicine SyndromeInternational Institute for Translational Chinese MedicineGuangzhou University of Chinese MedicineGuangzhou510006China
- Chinese Medicine Guangdong LaboratoryHengqinGuangdongChina
| | - Yuanhang Xu
- State Key Laboratory of Traditional Chinese Medicine SyndromeInternational Institute for Translational Chinese MedicineGuangzhou University of Chinese MedicineGuangzhou510006China
- Chinese Medicine Guangdong LaboratoryHengqinGuangdongChina
| | - Bingxin Wu
- State Key Laboratory of Traditional Chinese Medicine SyndromeInternational Institute for Translational Chinese MedicineGuangzhou University of Chinese MedicineGuangzhou510006China
- Chinese Medicine Guangdong LaboratoryHengqinGuangdongChina
| | - Jiachan Long
- State Key Laboratory of Traditional Chinese Medicine SyndromeInternational Institute for Translational Chinese MedicineGuangzhou University of Chinese MedicineGuangzhou510006China
- Chinese Medicine Guangdong LaboratoryHengqinGuangdongChina
| | - Caihong Liu
- State Key Laboratory of Traditional Chinese Medicine SyndromeInternational Institute for Translational Chinese MedicineGuangzhou University of Chinese MedicineGuangzhou510006China
- Chinese Medicine Guangdong LaboratoryHengqinGuangdongChina
| | - Zuhui Liang
- State Key Laboratory of Traditional Chinese Medicine SyndromeInternational Institute for Translational Chinese MedicineGuangzhou University of Chinese MedicineGuangzhou510006China
- Chinese Medicine Guangdong LaboratoryHengqinGuangdongChina
| | - Rong Zhang
- State Key Laboratory of Traditional Chinese Medicine SyndromeInternational Institute for Translational Chinese MedicineGuangzhou University of Chinese MedicineGuangzhou510006China
- Chinese Medicine Guangdong LaboratoryHengqinGuangdongChina
| | - Zhongqiu Liu
- State Key Laboratory of Traditional Chinese Medicine SyndromeInternational Institute for Translational Chinese MedicineGuangzhou University of Chinese MedicineGuangzhou510006China
- Chinese Medicine Guangdong LaboratoryHengqinGuangdongChina
| | - Caiyan Wang
- State Key Laboratory of Traditional Chinese Medicine SyndromeInternational Institute for Translational Chinese MedicineGuangzhou University of Chinese MedicineGuangzhou510006China
- Chinese Medicine Guangdong LaboratoryHengqinGuangdongChina
| |
Collapse
|
10
|
Brennan PN, MacMillan M, Manship T, Moroni F, Glover A, Troland D, MacPherson I, Graham C, Aird R, Semple SIK, Morris DM, Fraser AR, Pass C, McGowan NWA, Turner ML, Manson L, Lachlan NJ, Dillon JF, Kilpatrick AM, Campbell JDM, Fallowfield JA, Forbes SJ. Autologous macrophage therapy for liver cirrhosis: a phase 2 open-label randomized controlled trial. Nat Med 2025; 31:979-987. [PMID: 39794616 PMCID: PMC11922741 DOI: 10.1038/s41591-024-03406-8] [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: 06/05/2024] [Accepted: 11/11/2024] [Indexed: 01/13/2025]
Abstract
Cirrhosis is a major cause of morbidity and mortality; however, there are no approved therapies except orthotopic liver transplantation. Preclinical studies showed that bone-marrow-derived macrophage injections reduce inflammation, resolve fibrosis and stimulate liver regeneration. In a multicenter, open-label, parallel-group, phase 2 randomized controlled trial ( ISRCTN10368050 ) in n = 51 adult patients with compensated cirrhosis and Model for End-Stage Liver Disease (MELD) score ≥10 and ≤17, we evaluated the efficacy of autologous monocyte-derived macrophage therapy (n = 27) compared to standard medical care (n = 24). The primary endpoint was the difference in baseline to day 90 change in MELD score (ΔMELD) between treatment and control groups (ΔΔMELD). Secondary endpoints included adverse clinical outcomes, non-invasive fibrosis biomarkers and health-related quality of life (HRQoL) at 90 d, 180 d and 360 d. The ΔΔMELD between day 0 and day 90 in the treatment group compared to controls was -0.87 (95% confidence interval: -1.79, 0.0; P = 0.06); therefore, the primary endpoint was not met. During 360-d follow-up, five of 24 participants in the control group developed a total of 10 severe adverse events, four of which were liver related, and three deaths (two liver related), whereas no liver-related severe adverse events or deaths occurred in the treatment group. Although no differences were observed in biomarkers or HRQoL, exploratory analysis showed anti-inflammatory serum cytokine profiles after macrophage infusion. This study reinforces the safety and potential efficacy of macrophage therapy in cirrhosis, supporting further investigation.
Collapse
Affiliation(s)
- Paul N Brennan
- Division of Molecular and Clinical Medicine, University of Dundee, Dundee, UK
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Mark MacMillan
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Thomas Manship
- Edinburgh Transplant Centre, Royal Infirmary of Edinburgh, Edinburgh, UK
| | | | - Alison Glover
- Scottish National Blood Transfusion Service (SNBTS), Edinburgh, UK
| | - Debbie Troland
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Iain MacPherson
- Division of Molecular and Clinical Medicine, University of Dundee, Dundee, UK
| | - Catriona Graham
- Wellcome Trust Clinical Research Facility, University of Edinburgh, Edinburgh, UK
| | - Rhona Aird
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Scott I K Semple
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - David M Morris
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | | | - Chloe Pass
- Scottish National Blood Transfusion Service (SNBTS), Edinburgh, UK
| | - Neil W A McGowan
- Scottish National Blood Transfusion Service (SNBTS), Edinburgh, UK
| | - Marc L Turner
- Scottish National Blood Transfusion Service (SNBTS), Edinburgh, UK
| | - Lynn Manson
- Scottish National Blood Transfusion Service (SNBTS), Edinburgh, UK
| | | | - John F Dillon
- Division of Molecular and Clinical Medicine, University of Dundee, Dundee, UK
| | - Alastair M Kilpatrick
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | | | - Jonathan A Fallowfield
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Stuart J Forbes
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK.
- Edinburgh Transplant Centre, Royal Infirmary of Edinburgh, Edinburgh, UK.
| |
Collapse
|
11
|
Kim S, Kim YK, Kim S, Choi YS, Lee I, Joo H, Kim J, Kwon M, Park S, Jo MK, Choi Y, D'Souza T, Jung JW, Zakhem E, Lenzini S, Woo J, Choi H, Park J, Park SY, Kim GB, Nam GH, Kim IS. Dual-mode action of scalable, high-quality engineered stem cell-derived SIRPα-extracellular vesicles for treating acute liver failure. Nat Commun 2025; 16:1903. [PMID: 39988725 PMCID: PMC11847939 DOI: 10.1038/s41467-025-57133-w] [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: 07/07/2024] [Accepted: 02/06/2025] [Indexed: 02/25/2025] Open
Abstract
Acute liver failure (ALF) is a life-threatening condition caused by rapid hepatocyte death and impaired liver regeneration. Here we show that extracellular vesicles engineered to express Signal Regulatory Protein Alpha (SIRP-EVs), produced via a scalable 3D bioreactor process with high yield and purity, exhibit significant therapeutic potential by targeting damaged cells and promoting tissue repair. SIRP-EVs block CD47, a crucial inhibitory signal on necroptotic cells, to enhance macrophage-mediated clearance of dying hepatocytes. They also deliver regenerative cargo from mesenchymal stem cells, reprogramming macrophages to support liver regeneration. In male animal models, SIRP-EVs significantly reduce liver injury markers and improve survival, demonstrating their dual-function therapeutic efficacy. By integrating the resolution of necroptosis with regenerative macrophage reprogramming, SIRP-EVs represent a promising platform for restoring liver function. These findings support the development of EV-based in vivo macrophage reprogramming therapies for ALF and other inflammation-driven diseases, paving the way for the clinical application of engineered EV therapeutics.
Collapse
Grants
- This research was funded by National Research Foundation of Korea (NRF) funded by the Ministry of Science and ICT (Grant Number: RS-2017-NR022964).
- This research was supported by SHIFTBIO INC., Korean Fund for Regenerative Medicine funded by Ministry of Science and ICT, and Ministry of Health and Welfare (Grant Number: 23C0111L1), a grant of the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (Grant Number: RS-2023-KH136648), and a grant of the BIG3 Project, funded by the Ministry of SMEs and Startups, Republic of Korea (Grant Number: RS-2022-TI022422).
- This research was supported by a grant of the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (Grant Number: RS-2023-KH136648; RS-2023-KH140007).
Collapse
Affiliation(s)
| | | | | | | | - Inkyu Lee
- SHIFTBIO INC, Seoul, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Hyemin Joo
- SHIFTBIO INC, Seoul, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | | | - Minjeong Kwon
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Seryoung Park
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Min Kyoung Jo
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | - Jiwan Woo
- Research Animal Resource Center, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Hongyoon Choi
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
- Portrai, Inc, Seoul, Republic of Korea
| | | | - Seung-Yoon Park
- Department of Biochemistry, School of Medicine, Dongguk University, Gyeongju, Republic of Korea
| | | | - Gi-Hoon Nam
- SHIFTBIO INC, Seoul, Republic of Korea.
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, Republic of Korea.
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea.
- Chemical and Biological Integrative Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea.
| |
Collapse
|
12
|
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
|
13
|
Li W, Chen L, Zhou Q, Huang T, Zheng W, Luo F, Luo ZG, Zhang J, Liu J. Liver macrophage-derived exosomal miRNA-342-3p promotes liver fibrosis by inhibiting HPCAL1 in stellate cells. Hum Genomics 2025; 19:9. [PMID: 39910671 PMCID: PMC11800645 DOI: 10.1186/s40246-025-00722-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 01/20/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND The progression of liver fibrosis involves complex interactions between hepatic stellate cells (HSCs) and multiple immune cells in the liver, including macrophages. However, the mechanism of exosomes in the crosstalk between liver macrophages and HSCs remains unclear. METHOD Exosomes were extracted from primary mouse macrophages and cultured with HSCs, and the differential expression of microRNAs was evaluated using high-throughput sequencing technology. The functions of miR-342-3p in exosomes were verified by qPCR and luciferase reporter gene experiments with HSCs. The function of the target gene Hippocalcin-like protein 1 (HPCAL1) in HSCs was verified by Western blotting, qPCR, cellular immunofluorescence and co-IP in vivo and in vitro. RESULTS We demonstrated that exosomal microRNA-342-3p derived from primary liver macrophages could activate HSCs by inhibiting the expression of HPCAL1 in HSCs. HPCAL1, which is a fibrogenesis suppressor, could inhibit TGF-β signaling in HSCs by regulating the ubiquitination of Smad2 through direct interactions with its EF-hand 4 domain. CONCLUSION This study reveals a previously unidentified profibrotic mechanism of crosstalk between macrophages and HSCs in the liver and suggests an attractive novel therapeutic strategy for treating fibroproliferative liver diseases.
Collapse
Affiliation(s)
- Wenshuai Li
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Lirong Chen
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Qi Zhou
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Tiansheng Huang
- Department of Digestive Diseases, Shanghai Guanghua Hospital of Integrated Traditional Chinese And Western Medicine, Shanghai, 200040, China
| | - Wanwei Zheng
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Feifei Luo
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Zhong Guang Luo
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Jun Zhang
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Jie Liu
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
14
|
Zhu S, Jiang L, Liu X, Chen C, Luo X, Jiang S, Yin J, Liu X, Wu Y. m6A demethylase Fto inhibited macrophage activation and glycolysis in diabetic nephropathy via m6A/Npas2/Hif-1α axis. FASEB J 2025; 39:e70332. [PMID: 39831513 PMCID: PMC11744739 DOI: 10.1096/fj.202403014r] [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: 11/28/2024] [Revised: 12/28/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025]
Abstract
Macrophage infiltration and activation is a key factor in the progression of diabetic nephropathy (DN). However, aerobic glycolysis induced by m6A methylation modification plays a key role in M1-type activation of macrophages, but the specific mechanism remains unclear in DN. In this study, the expression of m6A demethylase Fto in bone marrow derived macrophages and primary kidney macrophages from db/db mice. Loss and gain-of-function analysis of Fto were performed to assess the role of Fto in DN. Transcriptome and MeRIP-seq association analysis was performed to identified the target gene was Npas2. In this study, we found that demethylase Fto exhibits low expression in type 2 DN m6A modification of Npas2 mediated by Fto regulates macrophages M1-type activation and glucose metabolism reprogramming to participate in the process of DN. Furthermore, Fto reduces the m6A modification level of Npas2 in macrophages through a Prrc2a-dependent mechanism, and decreasing its stability. This process mediates inflammation and glycolysis in M1 macrophages by regulating the Hif-1α signaling pathway. Fto may act as a suppressor of M1 macrophages inflammation and glycolysis in DN through the m6A/Npas2/Hif-1α axis. This findings providing a new basis for the prevention and treatment of DN.
Collapse
Affiliation(s)
- Sai Zhu
- Department of NephropathyThe First Affiliated Hospital of Anhui Medical University, Anhui Medical UniversityHefeiAnhuiPeople's Republic of China
| | - Ling Jiang
- Department of NephropathyThe First Affiliated Hospital of Anhui Medical University, Anhui Medical UniversityHefeiAnhuiPeople's Republic of China
| | - Xinran Liu
- Department of NephropathyThe First Affiliated Hospital of Anhui Medical University, Anhui Medical UniversityHefeiAnhuiPeople's Republic of China
| | - Chaoyi Chen
- Department of NephropathyThe First Affiliated Hospital of Anhui Medical University, Anhui Medical UniversityHefeiAnhuiPeople's Republic of China
| | - Xiaomei Luo
- Department of NephropathyThe First Affiliated Hospital of Anhui Medical University, Anhui Medical UniversityHefeiAnhuiPeople's Republic of China
| | - Shan Jiang
- Department of NephropathyThe First Affiliated Hospital of Anhui Medical University, Anhui Medical UniversityHefeiAnhuiPeople's Republic of China
| | - Jiuyu Yin
- Department of NephropathyThe First Affiliated Hospital of Anhui Medical University, Anhui Medical UniversityHefeiAnhuiPeople's Republic of China
| | - Xueqi Liu
- Department of NephropathyThe First Affiliated Hospital of Anhui Medical University, Anhui Medical UniversityHefeiAnhuiPeople's Republic of China
| | - Yonggui Wu
- Department of NephropathyThe First Affiliated Hospital of Anhui Medical University, Anhui Medical UniversityHefeiAnhuiPeople's Republic of China
- Center for Scientific Research of Anhui Medical UniversityHefeiAnhuiPeople's Republic of China
| |
Collapse
|
15
|
Candela ME, Addison M, Aird R, Man TY, Cartwright JA, Ashmore-Harris C, Kilpatrick AM, Starkey Lewis PJ, Drape A, Barnett M, Mitchell D, McLean C, McGowan N, Turner M, Dear JW, Forbes SJ. Cryopreserved human alternatively activated macrophages promote resolution of acetaminophen-induced liver injury in mouse. NPJ Regen Med 2025; 10:5. [PMID: 39843512 PMCID: PMC11754469 DOI: 10.1038/s41536-025-00393-3] [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: 06/24/2024] [Accepted: 01/10/2025] [Indexed: 01/24/2025] Open
Abstract
Acute liver failure is a rapidly progressing, life-threatening condition most commonly caused by an overdose of acetaminophen (paracetamol). The antidote, N-acetylcysteine (NAC), has limited efficacy when liver injury is established. If acute liver damage is severe, liver failure can rapidly develop with associated high mortality rates. We have previously demonstrated that alternatively, activated macrophages are a potential therapeutic option to reverse acute liver injury in pre-clinical models. In this paper, we present data using cryopreserved human alternatively activated macrophages (hAAMs)-which represent a potential, rapidly available treatment suitable for use in the acute setting. In a mouse model of APAP-induced injury, peripherally injected cryopreserved hAAMs reduced liver necrosis, modulated inflammatory responses, and enhanced liver regeneration. hAAMs were effective even when administered after the therapeutic window for NAC. This cell therapy approach represents a potential treatment for APAP overdose when NAC is ineffective because liver injury is established.
Collapse
Affiliation(s)
- Maria Elena Candela
- Centre for Regenerative Medicine, The Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK.
| | - Melisande Addison
- Centre for Regenerative Medicine, The Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Rhona Aird
- Centre for Regenerative Medicine, The Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Tak-Yung Man
- Centre for Regenerative Medicine, The Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Jennifer A Cartwright
- Centre for Regenerative Medicine, The Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
- The Royal (Dick) School of Veterinary Studies and the Roslin Institute, University of Edinburgh, Edinburgh, UK
| | - Candice Ashmore-Harris
- Centre for Regenerative Medicine, The Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Alastair M Kilpatrick
- Centre for Regenerative Medicine, The Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Philip J Starkey Lewis
- Centre for Regenerative Medicine, The Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Anna Drape
- Scottish National Blood Transfusion Service (SNBTS), The Jack Copland Centre, Heriot-Watt Research Park, Edinburgh, UK
| | - Mark Barnett
- Scottish National Blood Transfusion Service (SNBTS), The Jack Copland Centre, Heriot-Watt Research Park, Edinburgh, UK
| | - Donna Mitchell
- Scottish National Blood Transfusion Service (SNBTS), The Jack Copland Centre, Heriot-Watt Research Park, Edinburgh, UK
| | - Colin McLean
- Scottish National Blood Transfusion Service (SNBTS), The Jack Copland Centre, Heriot-Watt Research Park, Edinburgh, UK
| | - Neil McGowan
- Scottish National Blood Transfusion Service (SNBTS), The Jack Copland Centre, Heriot-Watt Research Park, Edinburgh, UK
| | - Marc Turner
- Scottish National Blood Transfusion Service (SNBTS), The Jack Copland Centre, Heriot-Watt Research Park, Edinburgh, UK
- Centre for Precision Cell Therapy for the Liver, Lothian Health Board, Queens Medical Research Institute, Edinburgh, UK
| | - James W Dear
- Centre for Precision Cell Therapy for the Liver, Lothian Health Board, Queens Medical Research Institute, Edinburgh, UK
- Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Stuart J Forbes
- Centre for Regenerative Medicine, The Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
- Centre for Precision Cell Therapy for the Liver, Lothian Health Board, Queens Medical Research Institute, Edinburgh, UK
| |
Collapse
|
16
|
Humphries C, Addison ML, Dear JW, Forbes SJ. The emerging role of alternatively activated macrophages to treat acute liver injury. Arch Toxicol 2025; 99:103-114. [PMID: 39503878 PMCID: PMC11742291 DOI: 10.1007/s00204-024-03892-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/09/2024] [Indexed: 01/19/2025]
Abstract
Acute liver injury (ALI) has a clear requirement for novel therapies. One emerging option is the use of alternatively activated macrophages (AAMs); a distinct subtype of macrophage with a role in liver injury control and repair. In this comprehensive review, we provide an overview of the current limited options for ALI, and the potential advantages offered by AAMs. We describe the evidence supporting their use from in vitro studies, pre-clinical animal studies, and human clinical trials. We suggest why the first evidence for the clinical use of AAMs is likely to be found in acetaminophen toxicity, and discuss the specific evidence for AAM use in this population, as well as potential applications for AAMs in other patient populations. The key domains by which the performance of AAMs for the treatment of ALI will be assessed are identified, and remaining challenges to the successful delivery of AAMs to clinic are explored.
Collapse
Affiliation(s)
- Chris Humphries
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Drive, Edinburgh, UK
| | - Melisande L Addison
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Drive, Edinburgh, UK
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - James W Dear
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Drive, Edinburgh, UK
| | - Stuart J Forbes
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK.
| |
Collapse
|
17
|
Qian Y, Zhao J, Wu H, Kong X. Innate immune regulation in inflammation resolution and liver regeneration in drug-induced liver injury. Arch Toxicol 2025; 99:115-126. [PMID: 39395921 DOI: 10.1007/s00204-024-03886-0] [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: 08/25/2024] [Accepted: 10/02/2024] [Indexed: 10/14/2024]
Abstract
Drug-induced liver injury (DILI) is an acute liver injury that poses a significant threat to human health. In severe cases, it can progress into chronic DILI or even lead to liver failure. DILI is typically caused by either intrinsic hepatotoxicity or idiosyncratic metabolic or immune responses. In addition to the direct damage drugs inflict on hepatocytes, the immune responses and liver inflammation triggered by hepatocyte death can further exacerbate DILI. Initially, we briefly discussed the differences in immune cell activation based on the type of liver cell death (hepatocytes, cholangiocytes, and LSECs). We then focused on the role of various immune cells (including macrophages, monocytes, neutrophils, dendritic cells, liver sinusoidal endothelial cells, eosinophils, natural killer cells, and natural killer T cells) in both the liver injury and liver regeneration stages of DILI. This article primarily reviews the role of innate immune regulation mediated by these immune cells in resolving inflammation and promoting liver regeneration during DILI, as well as therapeutic approaches targeting these immune cells for the treatment of DILI. Finally, we discussed the activation and function of liver progenitor cells (LPCs) during APAP-induced massive hepatic necrosis and the involvement of chronic inflammation in DILI.
Collapse
Affiliation(s)
- Yihan Qian
- Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Shanghai, China
| | - Jie Zhao
- Department of Liver Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hailong Wu
- Shanghai Key Laboratory of Molecular Imaging, Collaborative Innovation Center for Biomedicines, Shanghai University of Medicine and Health Sciences, Shanghai, China.
| | - Xiaoni Kong
- Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Shanghai, China.
| |
Collapse
|
18
|
Liu Y, Ye J, Fan Z, Wu X, Zhang Y, Yang R, Jiang B, Wang Y, Wu M, Zhou J, Meng J, Ge Z, Zhou G, Zhu Y, Xiao Y, Zheng M, Zhang S. Ginkgetin Alleviates Inflammation and Senescence by Targeting STING. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407222. [PMID: 39558862 PMCID: PMC11727237 DOI: 10.1002/advs.202407222] [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: 06/27/2024] [Revised: 09/27/2024] [Indexed: 11/20/2024]
Abstract
Ginkgo biloba extract is reported to have therapeutic effects on aging-related disorders. However, the specific component responsible for this biological function and its mechanism of action remain largely unknown. This study finds that Ginkgetin, an active ingredient of Ginkgo biloba extract, can alleviate cellular senescence and improve pathologies in multiple tissues of aging mice. To reveal the molecular mechanism of Ginkgetin's anti-aging effect, a graph convolutional network-based drug "on-target" pathway prediction algorithm for prediction is employed. The results indicate that the cGAS-STING pathway may be a potential target for Ginkgetin. Subsequent cell biological and biophysical data confirmed that Ginkgetin directly binds to the carboxy-terminal domain of STING protein, thereby inhibiting STING activation and signal transduction. Furthermore, in vivo pharmacodynamic data showed that Ginkgetin effectively alleviates systemic inflammation in Trex1-/- mice and inhibits the abnormally activated STING signaling in aging mouse model. In summary, this study, utilizing an artificial intelligence algorithm combined with pharmacological methods, confirms STING serves as a critical target for Ginkgetin in alleviating inflammation and senescence. Importantly, this study elucidates the specific component and molecular mechanism underlying the anti-aging effect of Ginkgo biloba extract, providing a robust theoretical basis for its therapeutic use.
Collapse
Affiliation(s)
- Yadan Liu
- Drug Discovery and Design CenterState Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- School of Chinese Materia MedicaNanjing University of Chinese MedicineNanjing210023China
| | - Jialin Ye
- CAS Key Laboratory of Tissue Microenvironment and TumorShanghai Institute of Nutrition and HealthChinese Academy of SciencesShanghai200031China
- University of Chinese Academy of SciencesBeijing100049China
| | - Zisheng Fan
- Shanghai Institute for Advanced Immunochemical StudiesSchool of Life Science and TechnologyShanghaiTech UniversityShanghai201210China
- Lingang LaboratoryShanghai200031China
| | - Xiaolong Wu
- Drug Discovery and Design CenterState Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- School of PharmacyEast China University of Science and TechnologyShanghai200237China
| | - Yinghui Zhang
- Drug Discovery and Design CenterState Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- University of Chinese Academy of SciencesBeijing100049China
| | - Ruirui Yang
- Drug Discovery and Design CenterState Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- University of Chinese Academy of SciencesBeijing100049China
| | - Bing Jiang
- Drug Discovery and Design CenterState Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- School of Chinese Materia MedicaNanjing University of Chinese MedicineNanjing210023China
| | - Yajie Wang
- Drug Discovery and Design CenterState Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- School of Pharmacology Science and TechnologyHangzhou Institute for Advanced StudyUniversity of Chinese Academy of SciencesHangzhou310024China
| | - Min Wu
- Drug Discovery and Design CenterState Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - Jingyi Zhou
- Drug Discovery and Design CenterState Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - Jingyi Meng
- Drug Discovery and Design CenterState Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- School of Chinese Materia MedicaNanjing University of Chinese MedicineNanjing210023China
| | - Zhiming Ge
- Drug Discovery and Design CenterState Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- School of Pharmacology Science and TechnologyHangzhou Institute for Advanced StudyUniversity of Chinese Academy of SciencesHangzhou310024China
| | - Guizhen Zhou
- Drug Discovery and Design CenterState Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- Shanghai Institute for Advanced Immunochemical StudiesSchool of Life Science and TechnologyShanghaiTech UniversityShanghai201210China
| | - Yuan Zhu
- Drug Discovery and Design CenterState Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- School of Chinese Materia MedicaNanjing University of Chinese MedicineNanjing210023China
| | - Yichuan Xiao
- CAS Key Laboratory of Tissue Microenvironment and TumorShanghai Institute of Nutrition and HealthChinese Academy of SciencesShanghai200031China
- University of Chinese Academy of SciencesBeijing100049China
| | - Mingyue Zheng
- Drug Discovery and Design CenterState Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- School of Chinese Materia MedicaNanjing University of Chinese MedicineNanjing210023China
- University of Chinese Academy of SciencesBeijing100049China
- Shanghai Institute for Advanced Immunochemical StudiesSchool of Life Science and TechnologyShanghaiTech UniversityShanghai201210China
- Lingang LaboratoryShanghai200031China
- School of Pharmacology Science and TechnologyHangzhou Institute for Advanced StudyUniversity of Chinese Academy of SciencesHangzhou310024China
| | - Sulin Zhang
- Drug Discovery and Design CenterState Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- University of Chinese Academy of SciencesBeijing100049China
| |
Collapse
|
19
|
Wang R, Chen Y, Han J, Ye H, Yang H, Li Q, He Y, Ma B, Zhang J, Ge Y, Wang Z, Sun B, Liu H, Cheng L, Wang Z, Lin G. Selectively targeting the AdipoR2-CaM-CaMKII-NOS3 axis by SCM-198 as a rapid-acting therapy for advanced acute liver failure. Nat Commun 2024; 15:10690. [PMID: 39681560 PMCID: PMC11649909 DOI: 10.1038/s41467-024-55295-7] [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: 04/13/2024] [Accepted: 12/08/2024] [Indexed: 12/18/2024] Open
Abstract
Acute liver failure (ALF) is a hepatology emergency with rapid hepatic destruction, multiple organ failures, and high mortality. Despite decades of research, established ALF has minimal therapeutic options. Here, we report that the small bioactive compound SCM-198 increases the survival of male ALF mice to 100%, even administered 24 hours after ALF establishment. We identify adiponectin receptor 2 (AdipoR2) as a selective target of SCM-198, with the AdipoR2 R335 residue being critical for the binding and signaling of SCM-198-AdipoR2 and AdipoR2 Y274 residue serving as a molecular switch for Ca2+ influx. SCM-198-AdipoR2 binding causes Ca2+ influx and elevates the phosphorylation levels of CaMKII and NOS3 in the AdipoR2-CaM-CaMKII-NOS3 complex identified in this study, rapidly inducing nitric oxide production for liver protection in murine ALF. SCM-198 also protects human ESC-derived liver organoids from APAP/TAA injuries. Thus, selectively targeting the AdipoR2-CaM-CaMKII-NOS3 axis by SCM-198 is a rapid-acting therapeutic strategy for advanced ALF.
Collapse
Affiliation(s)
- Rui Wang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedic, Tongji Hospital affiliated to Tongji University, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Youwei Chen
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedic, Tongji Hospital affiliated to Tongji University, School of Life Sciences and Technology, Tongji University, Shanghai, China
- School of Medicine, Tongji University, Shanghai, China
| | - Jiazhen Han
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedic, Tongji Hospital affiliated to Tongji University, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Huikang Ye
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedic, Tongji Hospital affiliated to Tongji University, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Huiran Yang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedic, Tongji Hospital affiliated to Tongji University, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Qianyan Li
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedic, Tongji Hospital affiliated to Tongji University, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yizhen He
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedic, Tongji Hospital affiliated to Tongji University, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Boyu Ma
- Department of Gastroenterology, Tongji Hospital affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, China
| | - Junjie Zhang
- Department of Gastroenterology, Tongji Hospital affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, China
| | - Yanli Ge
- Department of Gastroenterology, Tongji Hospital affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, China
| | - Zhe Wang
- Department of Gastroenterology, Tongji Hospital affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, China
| | - Bo Sun
- Department of Gastroenterology, Tongji Hospital affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, China
| | - Huahua Liu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedic, Tongji Hospital affiliated to Tongji University, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Liming Cheng
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedic, Tongji Hospital affiliated to Tongji University, School of Life Sciences and Technology, Tongji University, Shanghai, China.
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, China.
| | - Zhirong Wang
- Department of Gastroenterology, Tongji Hospital affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, China.
| | - Gufa Lin
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedic, Tongji Hospital affiliated to Tongji University, School of Life Sciences and Technology, Tongji University, Shanghai, China.
- School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
20
|
Humphries C, Addison M, Aithal G, Boyd J, Briody L, Campbell JDM, Candela ME, Clarke E, Coulson J, Downing-James N, Fontana RJ, Geddes A, Grahamslaw J, Grant A, Heye A, Hutchinson JA, Jones A, Mitchell F, Moore J, Riddell A, Rodriguez A, Thomas A, Tucker G, Walker K, Weir CJ, Woods R, Zahra S, Forbes SJ, Dear JW. Macrophage Therapy for Acute Liver Injury (MAIL): a study protocol for a phase 1 randomised, open-label, dose-escalation study to evaluate safety, tolerability and activity of allogeneic alternatively activated macrophages in patients with paracetamol-induced acute liver injury in the UK. BMJ Open 2024; 14:e089417. [PMID: 39653576 PMCID: PMC11628987 DOI: 10.1136/bmjopen-2024-089417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 11/04/2024] [Indexed: 12/12/2024] Open
Abstract
INTRODUCTION Acute liver failure (ALF) has no effective treatment other than liver transplantation and is commonly caused by paracetamol overdose. New treatments are needed to treat and prevent ALF. Alternatively-activated macrophages (AAMs) can promote resolution of liver necrosis and stimulate hepatocyte proliferation. Using AAMs in unscheduled care requires the use of an allogeneic product. A clinical trial is needed to determine the safety and tolerability of allogeneic AAMs. METHODS AND ANALYSIS A single-centre, open-label, dose-escalation, phase 1 randomised trial to determine whether there is dose-limiting toxicity of AAMs in patients with paracetamol-induced acute liver injury. Randomisation will occur at higher doses. Between 17 and 30 patients will receive treatment, subject to dose-limiting toxicity and an adaptive trial design which aims to reduce the risk of allocation bias through blinding and randomisation. ETHICS AND DISSEMINATION The trial will be conducted according to the ethical principles of the Declaration of Helsinki 2013 and has been approved by North East-York Research Ethics Committee (reference 23/NE/0019), National Health Service Lothian Research and Development department, and the UK Medicines and Healthcare products Regulatory Agency. When the trial concludes, results will be shared by presentation and publication. TRIAL REGISTRATION NUMBER ISRCTN12637839.
Collapse
Affiliation(s)
- Christopher Humphries
- Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, UK
| | - Melisande Addison
- Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, UK
| | - Guruprasad Aithal
- MAIL Trial Data Monitoring Committee, Edinburgh Clinical Trials Unit, Usher Institute, University of Edinburgh, 3 Little France Road, Edinburgh, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust, Queen's Medical Centre, Derby Road, Nottingham, UK
| | - Julia Boyd
- Edinburgh Clinical Trials Unit, Usher Institute, University of Edinburgh, 3 Little France Road, Edinburgh, UK
| | - Lesley Briody
- Edinburgh Clinical Research Facility, Royal Infirmary of Edinburgh, 51 Little France Crescent, Edinburgh, UK
| | - John D M Campbell
- Scottish National Blood Transfusion Service, Jack Copland Centre, 52 Research Avenue North, Edinburgh, UK
| | - Maria Elena Candela
- Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, UK
| | - Ellise Clarke
- Emergency Medicine Research Group Edinburgh, Royal Infirmary of Edinburgh, 51 Little France Crescent, Edinburgh, UK
| | - James Coulson
- MAIL Trial Data Monitoring Committee, Edinburgh Clinical Trials Unit, Usher Institute, University of Edinburgh, 3 Little France Road, Edinburgh, UK
| | - Nicholas Downing-James
- Edinburgh Clinical Research Facility, Royal Infirmary of Edinburgh, 51 Little France Crescent, Edinburgh, UK
| | - Robert John Fontana
- MAIL Trial Steering Committee, Edinburgh Clinical Trials Unit, Usher Institute, University of Edinburgh, 3 Little France Road, Edinburgh, UK
| | - Ailsa Geddes
- Edinburgh Clinical Research Facility, Royal Infirmary of Edinburgh, 51 Little France Crescent, Edinburgh, UK
| | - Julia Grahamslaw
- Emergency Medicine Research Group Edinburgh, Royal Infirmary of Edinburgh, 51 Little France Crescent, Edinburgh, UK
| | - Alison Grant
- Emergency Medicine Research Group Edinburgh, Royal Infirmary of Edinburgh, 51 Little France Crescent, Edinburgh, UK
| | - Anna Heye
- Edinburgh Clinical Trials Unit, Usher Institute, University of Edinburgh, 3 Little France Road, Edinburgh, UK
| | - James A Hutchinson
- MAIL Trial Steering Committee, Edinburgh Clinical Trials Unit, Usher Institute, University of Edinburgh, 3 Little France Road, Edinburgh, UK
| | - Ashley Jones
- MAIL Trial Data Monitoring Committee, Edinburgh Clinical Trials Unit, Usher Institute, University of Edinburgh, 3 Little France Road, Edinburgh, UK
| | - Fiona Mitchell
- Edinburgh Clinical Research Facility, Royal Infirmary of Edinburgh, 51 Little France Crescent, Edinburgh, UK
| | - Joanna Moore
- MAIL Trial Data Monitoring Committee, Edinburgh Clinical Trials Unit, Usher Institute, University of Edinburgh, 3 Little France Road, Edinburgh, UK
| | - Alice Riddell
- MAIL Trial Steering Committee, Edinburgh Clinical Trials Unit, Usher Institute, University of Edinburgh, 3 Little France Road, Edinburgh, UK
| | - Aryelly Rodriguez
- Edinburgh Clinical Trials Unit, Usher Institute, University of Edinburgh, 3 Little France Road, Edinburgh, UK
| | - Angela Thomas
- MAIL Trial Steering Committee, Edinburgh Clinical Trials Unit, Usher Institute, University of Edinburgh, 3 Little France Road, Edinburgh, UK
| | - Garry Tucker
- Edinburgh Clinical Research Facility, Royal Infirmary of Edinburgh, 51 Little France Crescent, Edinburgh, UK
| | - Kim Walker
- Edinburgh Clinical Research Facility, Royal Infirmary of Edinburgh, 51 Little France Crescent, Edinburgh, UK
| | - Christopher J Weir
- Edinburgh Clinical Trials Unit, Usher Institute, University of Edinburgh, 3 Little France Road, Edinburgh, UK
| | - Rachel Woods
- Edinburgh Clinical Research Facility, Royal Infirmary of Edinburgh, 51 Little France Crescent, Edinburgh, UK
| | - Sharon Zahra
- Scottish National Blood Transfusion Service, Jack Copland Centre, 52 Research Avenue North, Edinburgh, UK
| | - Stuart J Forbes
- Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, UK
- Centre for Precision Cell Therapy for the Liver, Lothian Health Board, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, UK
| | - James W Dear
- Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, UK
- Centre for Precision Cell Therapy for the Liver, Lothian Health Board, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, UK
| |
Collapse
|
21
|
Wang Q, Wang F, Zhou Y, Li X, Xu S, Tang L, Jin Q, Fu A, Yang R, Li W. Bacillus amyloliquefaciens SC06 Attenuated Lipopolysaccharide-Induced acute liver injury by suppressing bile acid-associated NLRP3 inflammasome activation. Int Immunopharmacol 2024; 142:113129. [PMID: 39293317 DOI: 10.1016/j.intimp.2024.113129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 09/02/2024] [Accepted: 09/06/2024] [Indexed: 09/20/2024]
Abstract
The involvement of the inflammatory response has been linked to the development of liver illnesses. As medications with the potential to prevent and cure liver illness, probiotics have garnered an increasing amount of interest in recent years. The present study used a piglet model with acute liver injury (ALI) induced by lipopolysaccharides (LPS) to investigate the regulatory mechanisms of Bacillus amyloliquefaciens SC06. Our findings indicated that SC06 mitigated the liver structural damage caused by LPS, as shown by the decreased infiltration of inflammatory cells and the enhanced structural integrity. In addition, After the administration of SC06, there was a reduction in the increased levels of the liver damage markers. In the LPS group, there was an increase in the mRNA expression of inflammatory cytokines, apoptosis cell rate, and genes associated with apoptosis, while these alterations were mitigated by SC06 administration. Furthermore, SC06 prevented pigs from suffering liver damage by preventing the activation of the NLRP3 inflammasome, which was normally triggered by LPS. The examination of serum metabolic pathways found that ALI was related to several metabolic processes, including primary bile acid biosynthesis, pentose and glucuronate interconversions and the metabolism of phenylalanine. Significantly, our research revealed that the administration of SC06 effectively controlled the concentrations of bile acids in the serum. The correlation results also revealed clear relationships between bile acids and liver characteristics and NLRP3 inflammasome-related genes. However, in vitro experiments revealed that SC06 could not directly inhibit NLRP3 activation under ATP, monosodium urate, and nigericin stimulation, while taurochenodeoxycholic acid (TCDCA) activated NLRP3 inflammasome related genes. In conclusion, our study proved that the hepaprotective effect of SC06 on liver injury, which was closely associated with the restoration of bile acids homeostasis and NLRP3 inflammasome inhibition.
Collapse
Affiliation(s)
- Qi Wang
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fei Wang
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yuanhao Zhou
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiang Li
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shujie Xu
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Li Tang
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qian Jin
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Aikun Fu
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Rongchang Yang
- School of Veterinary and Life Sciences, Murdoch University, Murdoch, Western Australia, 6150, Australia; Zhejiang Youheyhey Biotechnology Co., LTD, Huzhou 313000, Zhejiang Province, China.
| | - Weifen Li
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
22
|
Zhang L, Deng Y, Bai X, Wei X, Ren Y, Chen S, Deng H. Cell therapy for end-stage liver disease: Current state and clinical challenge. Chin Med J (Engl) 2024; 137:2808-2820. [PMID: 39602326 DOI: 10.1097/cm9.0000000000003332] [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: 06/08/2024] [Indexed: 11/29/2024] Open
Abstract
ABSTRACT Liver disease involves a complex interplay of pathological processes, including inflammation, hepatocyte necrosis, and fibrosis. End-stage liver disease (ESLD), such as liver failure and decompensated cirrhosis, has a high mortality rate, and liver transplantation is the only effective treatment. However, to overcome problems such as the shortage of donor livers and complications related to immunosuppression, there is an urgent need for new treatment strategies that need to be developed for patients with ESLD. For instance, hepatocytes derived from donor livers or stem cells can be engrafted and multiplied in the liver, substituting the host hepatocytes and rebuilding the liver parenchyma. Stem cell therapy, especially mesenchymal stem cell therapy, has been widely proved to restore liver function and alleviate liver injury in patients with severe liver disease, which has contributed to the clinical application of cell therapy. In this review, we discussed the types of cells used to treat ESLD and their therapeutic mechanisms. We also summarized the progress of clinical trials around the world and provided a perspective on cell therapy.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | | | | | | | | | | | | |
Collapse
|
23
|
Ma X, Qiu J, Zou S, Tan L, Miao T. The role of macrophages in liver fibrosis: composition, heterogeneity, and therapeutic strategies. Front Immunol 2024; 15:1494250. [PMID: 39635524 PMCID: PMC11616179 DOI: 10.3389/fimmu.2024.1494250] [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: 09/10/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024] Open
Abstract
Macrophages, the predominant immune cells in the liver, are essential for maintaining hepatic homeostasis and responding to liver injury caused by external stressors. The hepatic macrophage population is highly heterogeneous and plastic, mainly comprised of hepatic resident kuffer cells (KCs), monocyte-derived macrophages (MoMφs), lipid-associated macrophages (LAMs), and liver capsular macrophages (LCMs). KCs, a population of resident macrophages, are localized in the liver and can self-renew through in situ proliferation. However, MoMφs in the liver are recruited from the periphery circulation. LAMs are a self-renewing subgroup of liver macrophages near the bile duct. While LCMs are located in the liver capsule and derived from peripheral monocytes. LAMs and LCMs are also involved in liver damage induced by various factors. Hepatic macrophages exhibit distinct phenotypes and functions depending on the specific microenvironment in the liver. KCs are critical for initiating inflammatory responses after sensing tissue damage, while the MoMφs infiltrated in the liver are implicated in both the progression and resolution of chronic hepatic inflammation and fibrosis. The regulatory function of liver macrophages in hepatic fibrosis has attracted significant interest in current research. Numerous literatures have documented that the MoMφs in the liver have a dual impact on the progression and resolution of liver fibrosis. The MoMφs in the liver can be categorized into two subtypes based on their Ly-6C expression level: inflammatory macrophages with high Ly-6C expression (referred to as Ly-6Chi subgroup macrophages) and reparative macrophages with low Ly-6C expression (referred to as Ly-6Clo subgroup macrophages). Ly-6Chi subgroup macrophages are conducive to the occurrence and progression of liver fibrosis, while Ly-6Clo subgroup macrophages are associated with the degradation of extracellular matrix (ECM) and regression of liver fibrosis. Given this, liver macrophages play a pivotal role in the occurrence, progression, and regression of liver fibrosis. Based on these studies, treatment therapies targeting liver macrophages are also being studied gradually. This review aims to summarize researches on the composition and origin of liver macrophages, the macrophage heterogeneity in the progression and regression of liver fibrosis, and anti-fibrosis therapeutic strategies targeting macrophages in the liver.
Collapse
Affiliation(s)
- Xiaocao Ma
- Department of Nuclear Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jia Qiu
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Intelligent Medical Imaging of Jiangxi Key Laboratory, Nanchang, China
| | - Shubiao Zou
- Department of Nuclear Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Liling Tan
- Department of Nuclear Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Tingting Miao
- Department of Nuclear Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
24
|
Chao S, Shan S, Liu Z, Liu Z, Wang S, Qiang Y, Ni W, Li H, Cheng D, Jia Q, Song F. Both TREM2-dependent macrophages and Kupffer cells play a protective role in APAP-induced acute liver injury. Int Immunopharmacol 2024; 141:112926. [PMID: 39159559 DOI: 10.1016/j.intimp.2024.112926] [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: 06/16/2024] [Revised: 07/22/2024] [Accepted: 08/09/2024] [Indexed: 08/21/2024]
Abstract
The inflammatory response is a significant factor in acetaminophen (APAP)-induced acute liver injury. And it can be mediated by macrophages of different origins. However, whether Kupffer cells and mononuclear-derived macrophages play an injury or protective role in APAP hepatotoxicity is still unclear. In this study, C57/BL6N mice were performed to establish the APAP acute liver injury model. Intervention experiments were also carried out using clodronate liposomes or TREM2 knockout. We found that APAP overdose triggered the activation of inflammatory factors and enhanced the expression of the RIPK1-MLKL pathway in mice's livers. Moreover, our study showed that inflammation-related protein expression was increased after clodronate liposome administration or TREM2 knockout. The RIPK1-MLKL-mediated necroptosis was also significantly activated after the elimination of Kupffer cells or the inhibition of mononuclear-derived macrophages. More importantly, clodronate liposomes treatment and TREM2 deficiency all worsen APAP-induced liver damage in mice. In conclusion, the results indicate that Kupffer cells and mononuclear macrophages play a protective role in APAP-induced liver injury by regulating necroptosis. Therefore, macrophages hold as a potential therapeutic target for APAP-induced liver damage.
Collapse
Affiliation(s)
- Shihua Chao
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, China; Qinghai Center for Disease Control and Prevention, No. 55, Bayi Middle Road, Chengdong District, Xining City, Qinghai Province 810000, China
| | - Shulin Shan
- Department of Health Test and Detection, Shandong Center for Disease Control and Prevention, 16992 Jingshi Road, Ji'nan, Shandong 250014, China
| | - Zhaoxiong Liu
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, China
| | - Zhidan Liu
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Science, Ji'nan 250062, China
| | - Shuai Wang
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, China
| | - Yalong Qiang
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, China
| | - Wenting Ni
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, China
| | - Hui Li
- Department of Health Test and Detection, Shandong Center for Disease Control and Prevention, 16992 Jingshi Road, Ji'nan, Shandong 250014, China
| | - Dong Cheng
- Department of Health Test and Detection, Shandong Center for Disease Control and Prevention, 16992 Jingshi Road, Ji'nan, Shandong 250014, China
| | - Qiang Jia
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Science, Ji'nan 250062, China
| | - Fuyong Song
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, China.
| |
Collapse
|
25
|
Zhu S, Chen X, Sun L, Li X, Chen Y, Li L, Suo X, Xu C, Ji M, Wang J, Wang H, Zhang L, Meng X, Huang C, Li J. N6-Methyladenosine modification of circDcbld2 in Kupffer cells promotes hepatic fibrosis via targeting miR-144-3p/Et-1 axis. Acta Pharm Sin B 2024. [DOI: 10.1016/j.apsb.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
|
26
|
Zhuang J, Zhang H, Wu J, Hu D, Meng T, Xue J, Xu H, Wang G, Wang H, Zhang G. Redox-Responsive AIEgen Diselenide-Covalent Organic Framework Composites Targeting Hepatic Macrophages for Treatment of Drug-induced Liver Injury. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402656. [PMID: 39140196 DOI: 10.1002/smll.202402656] [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: 04/03/2024] [Revised: 08/03/2024] [Indexed: 08/15/2024]
Abstract
The escalating misuse of antipyretic and analgesic drugs, alongside the rising incidents of acute drug-induced liver injury, underscores the need for a precisely targeted drug delivery system. Herein, two isoreticular covalent organic frameworks (Se-COF and Se-BCOF) are developed by Schiff-base condensation of emissive tetraphenylethylene and diselenide-bridged monomers. Leveraging the specific affinity of macrophages for mannose, the first precise targeting of these COFs to liver macrophages is achieved. The correlation is also explored between the therapeutic effects of COFs and the NLRP3/ASC/Caspase-1 signaling pathway. Utilizing this innovative delivery vehicle, the synergistic delivery of matrine and berberine are accomplished, compounds extracted from traditional Chinese medicine. This approach not only demonstrated the synergistic effects of the drugs but also mitigated their toxicity. Notably, berberine, through phosphorylation of JNK and up-regulation of nuclear Nrf-2 and its downstream gene Mn-SOD expression, simultaneously countered excessive ROS and suppressed the activation of the NLRP3/ASC/Caspase-1 signaling pathway in injured liver tissues. This multifaceted approach proved highly effective in safeguarding against acute drug-induced liver injury, ultimately restoring liver health to normalcy. These findings present a novel and promising strategy for the treatment of acute drug-induced liver injury.
Collapse
Affiliation(s)
- Jialu Zhuang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Hao Zhang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Jin Wu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Danyou Hu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Tao Meng
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Jing Xue
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Hanyang Xu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Gang Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Guiyang Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| |
Collapse
|
27
|
Ashmore-Harris C, Antonopoulou E, Aird RE, Man TY, Finney SM, Speel AM, Lu WY, Forbes SJ, Gadd VL, Waters SL. Utilising an in silico model to predict outcomes in senescence-driven acute liver injury. NPJ Regen Med 2024; 9:26. [PMID: 39349489 PMCID: PMC11442582 DOI: 10.1038/s41536-024-00371-1] [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: 09/29/2023] [Accepted: 09/17/2024] [Indexed: 10/02/2024] Open
Abstract
Currently liver transplantation is the only treatment option for liver disease, but organ availability cannot meet patient demand. Alternative regenerative therapies, including cell transplantation, aim to modulate the injured microenvironment from inflammation and scarring towards regeneration. The complexity of the liver injury response makes it challenging to identify suitable therapeutic targets when relying on experimental approaches alone. Therefore, we adopted a combined in vivo-in silico approach and developed an ordinary differential equation model of acute liver disease able to predict the host response to injury and potential interventions. The Mdm2fl/fl mouse model of senescence-driven liver injury was used to generate a quantitative dynamic characterisation of the key cellular players (macrophages, endothelial cells, myofibroblasts) and extra cellular matrix involved in liver injury. This was qualitatively captured by the mathematical model. The mathematical model was then used to predict injury outcomes in response to milder and more severe levels of senescence-induced liver injury and validated with experimental in vivo data. In silico experiments using the validated model were then performed to interrogate potential approaches to enhance regeneration. These predicted that increasing the rate of macrophage phenotypic switch or increasing the number of pro-regenerative macrophages in the system will accelerate the rate of senescent cell clearance and resolution. These results showcase the potential benefits of mechanistic mathematical modelling for capturing the dynamics of complex biological systems and identifying therapeutic interventions that may enhance our understanding of injury-repair mechanisms and reduce translational bottlenecks.
Collapse
Affiliation(s)
- Candice Ashmore-Harris
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh BioQuarter, Edinburgh, UK
| | | | - Rhona E Aird
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh BioQuarter, Edinburgh, UK
| | - Tak Yung Man
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh BioQuarter, Edinburgh, UK
| | - Simon M Finney
- Mathematical Institute, University of Oxford, Oxford, UK
| | - Annelijn M Speel
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh BioQuarter, Edinburgh, UK
| | - Wei-Yu Lu
- Centre for Inflammation Research, Institute for Regeneration & Repair, University of Edinburgh, Edinburgh BioQuarter, Edinburgh, UK
| | - Stuart J Forbes
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh BioQuarter, Edinburgh, UK
| | - Victoria L Gadd
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh BioQuarter, Edinburgh, UK.
| | - Sarah L Waters
- Mathematical Institute, University of Oxford, Oxford, UK.
| |
Collapse
|
28
|
Zhang X, Geng Q, Lin L, Zhang L, Shi C, Liu B, Yan L, Cao Z, Li L, Lu P, Tan Y, He X, Zhao N, Li L, Lu C. Insights gained into the injury mechanism of drug and herb induced liver injury in the hepatic microenvironment. Toxicology 2024; 507:153900. [PMID: 39079402 DOI: 10.1016/j.tox.2024.153900] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/17/2024]
Abstract
Drug-Induced Liver Injury (DILI) and herb Induced Liver Injury (HILI) continues to pose a substantial challenge in both clinical practice and drug development, representing a grave threat to patient well-being. This comprehensive review introduces a novel perspective on DILI and HILI by thoroughly exploring the intricate microenvironment of the liver. The dynamic interplay among hepatocytes, sinusoidal endothelial cells, Kupffer cells, hepatic stellate cells, cholangiocytes, and the intricate vascular network assumes a central role in drug metabolism and detoxification. Significantly, this microenvironment is emerging as a critical determinant of susceptibility to DILI and HILI. The review delves into the multifaceted interactions within the liver microenvironment, providing valuable insights into the complex mechanisms that underlie DILI and HILI. Furthermore, we discuss potential strategies for mitigating drug-induced liver injury by targeting these influential factors, emphasizing their clinical relevance. By highlighting recent advances and future prospects, our aim is to shed light on the promising avenue of leveraging the liver microenvironment for the prevention and mitigation of DILI and HILI. This deeper understanding is crucial for advancing clinical practices and ensuring patient safety in the realm of DILI and HILI.
Collapse
Affiliation(s)
- Xiaomeng Zhang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qi Geng
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lin Lin
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lulu Zhang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Changqi Shi
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bin Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lan Yan
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhiwen Cao
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Li
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Peipei Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yong Tan
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaojuan He
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ning Zhao
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Li
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
29
|
Torabi S, Zarrabi M, Shekari F, Poorkazem H, Lotfinia M, Bencina S, Gramignoli R, Hassan M, Najimi M, Vosough M. Wharton's Jelly mesenchymal stem cell-derived extracellular vesicles induce liver fibrosis-resolving phenotype in alternatively activated macrophages. J Cell Mol Med 2024; 28:e18507. [PMID: 39288445 PMCID: PMC11407755 DOI: 10.1111/jcmm.18507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 05/30/2024] [Accepted: 06/08/2024] [Indexed: 09/19/2024] Open
Abstract
The potential of extracellular vesicles (EVs) isolated from mesenchymal stromal cells in guiding macrophages toward anti-inflammatory immunophenotypes, has been reported in several studies. In our study, we provided experimental evidence of a distinctive effect played by Wharton Jelly mesenchymal stromal cell-derived EVs (WJ-EVs) on human macrophages. We particularly analyzed their anti-inflammatory effects on macrophages by evaluating their interactions with stellate cells, and their protective role in liver fibrosis. A three-step gradient method was used to isolate monocytes from umbilical cord blood (UCB). Two subpopulations of WJ-EVs were isolated by high-speed (20,000 g) and differential ultracentrifugation (110,000 g). Further to their characterization, they were designated as EV20K and EV110K and incubated at different concentrations with UCB-derived monocytes for 7 days. Their anti-fibrotic effect was assessed by studying the differentiation and functional levels of generated macrophages and their potential to modulate the survival and activity of LX2 stellate cells. The EV20K triggers the polarization of UCB-derived monocytes towards a peculiar M2-like functional phenotype more effectively than the M-CSF positive control. The EV20K treated macrophages were characterized by a higher expression of scavenger receptors, increased phagocytic capacity and production level of interleukin-10 and transforming growth factor-β. Conditioned medium from those polarized macrophages attenuated the proliferation, contractility and activation of LX2 stellate cells. Our data show that EV20K derived from WJ-MSCs induces activated macrophages to suppress immune responses and potentially play a protective role in the pathogenesis of liver fibrosis by directly inhibiting HSC's activation.
Collapse
Affiliation(s)
- Shukoofeh Torabi
- Department of Regenerative Medicine, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
- Department of Tissue Engineering and Regenerative Medicine, School of Advanced Technologies in MedicineMazandaran University of Medical SciencesSariIran
| | - Morteza Zarrabi
- Department of Regenerative Medicine, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
| | - Hedie Poorkazem
- Department of Stem Cells and Developmental Biology, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
| | - Majid Lotfinia
- Physiology Research CenterKashan University of Medical SciencesKashanIran
| | - Stefan Bencina
- Department of Laboratory Medicine, Division of PathologyKarolinska InstitutetStockholmSweden
| | - Roberto Gramignoli
- Department of Laboratory Medicine, Division of PathologyKarolinska InstitutetStockholmSweden
- UOSD Cell FactoryIRCCS Istituto Giannina GasliniGenoaItaly
| | - Moustapha Hassan
- Experimental Cancer Medicine, Institution for Laboratory Medicine and Karolinska University HospitalKarolinska InstituteStockholmSweden
| | - Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell TherapyInstitute of Experimental and Clinical Research (IREC), UCLouvainBrusselsBelgium
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
- Experimental Cancer Medicine, Institution for Laboratory Medicine and Karolinska University HospitalKarolinska InstituteStockholmSweden
| |
Collapse
|
30
|
Zhang L, Tan J. Effect of primary osteoblast-derived extracellular vesicles on osteoclast differentiation. Zhejiang Da Xue Xue Bao Yi Xue Ban 2024; 53:434-442. [PMID: 39034117 PMCID: PMC11375487 DOI: 10.3724/zdxbyxb-2024-0148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
OBJECTIVES To investigate the effect of osteoblast-derived extracellular vesicles (OB-EVs) on the proliferation and differentiation of osteoclasts, and to explore the possible molecular mechanism of extracellular vesicles involved in the communication between osteoblasts and osteoclasts. METHODS Primary osteoblasts were isolated from newborn mouse calvarial bone and induced by β-glycero phosphate, ascorbic acid and dexamethasone. Osteogenic feature was tested by alkaline phosphatase (ALP) and alizarin red S staining. Extracellular vesicles were isolated by ultracentrifugation from the cell culture supernatant. Vesicle morphology was observed by transmission electron microscopy, and the characteristic markers of tumor susceptibility gene 101 (TSG101), ALG-2 interacting protein X (Alix) and cluster of differentiation 9 (CD9) on the surface of extracellular vesicles were identified by Western blotting. Cell counting kit 8 (CCK-8) assay was used to determine the proliferation effect of OB-EVs on mouse mononuclear macrophage RAW264.7 cells. Furthermore, the expression level of specific markers of osteoclast differentiation in RAW264.7 cells was detected by Western blotting after the combined effect of OB-EVs and receptor activator for nuclear factor κB ligand (RANKL). The number of osteoclasts was observed and compared with OB-EVs-treated mouse bone marrow-derived macrophages (BMMs) by tartrate-resistant acid phosphatase (TRAP) staining, and the effect of OB-EVs on osteoclast differentiation was determined. RESULTS The extracted OB-EVs showed a double-layer cup-like structure with a diameter of 30-150 nm, and TSG101, Alix and CD9 were expressed. RAW264.7 cells were stimulated with OB-EVs, and the results of CCK-8 assay showed that high concentration of OB-EVs (more than 20 μg/mL) inhibited cell proliferation (P<0.05). Western blotting analysis showed that the expression of osteoclast differentiation marker proteins such as c-Fos, activated T cell nuclear factor (NFATc1) and c-Jun N-terminal kinase (JNK) in RAW264.7 cells were significantly increased, and the promoting effect was enhanced with increasing of OB-EVs concentration (P<0.05). In addition, the combination of OB-EVs and RANKL on BMMs showed that the number of TRAP-positive cells was significantly higher than that of the RANKL induction group alone (P<0.05). CONCLUSIONS OB-EVs can promote the differentiation of osteoclast precursor cells into osteoclasts, but high concentration of OB-EVs can inhibit proliferation of RAW264.7 cells.
Collapse
Affiliation(s)
- Lan Zhang
- Department of Stomatology, Zhejiang Hospital, Hangzhou 310030, China.
| | - Jingyi Tan
- Department of Stomatology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.
| |
Collapse
|
31
|
Liu J, Liu J, Mu W, Ma Q, Zhai X, Jin B, Liu Y, Zhang N. Delivery Strategy to Enhance the Therapeutic Efficacy of Liver Fibrosis via Nanoparticle Drug Delivery Systems. ACS NANO 2024; 18:20861-20885. [PMID: 39082637 DOI: 10.1021/acsnano.4c02380] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Liver fibrosis (LF) is a pathological repair reaction caused by a chronic liver injury that affects the health of millions of people worldwide, progressing to life-threatening cirrhosis and liver cancer without timely intervention. Due to the complexity of LF pathology, multiple etiological characteristics, and the deposited extracellular matrix, traditional drugs cannot reach appropriate targets in a time-space matching way, thus decreasing the therapeutic effect. Nanoparticle drug delivery systems (NDDS) enable multidrug co-therapy and develop multifactor delivery strategies targeting pathological processes, showing great potential in LF therapy. Based on the pathogenesis and the current clinical treatment status of LF, we systematically elucidate the targeting mechanism of NDDS used in the treatment of LF. Subsequently, we focus on the progress of drug delivery applications for LF, including combined delivery for the liver fibrotic pathological environment, overcoming biological barriers, precise intracellular regulation, and intelligent responsive delivery for the liver fibrotic microenvironment. We hope that this review will inspire the rational design of NDDS for LF in the future in order to provide ideas and methods for promoting LF regression and cure.
Collapse
Affiliation(s)
- Jie Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Jinhu Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Weiwei Mu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Qingping Ma
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xiangyu Zhai
- Department of Hepatobiliary Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, China
| | - Bin Jin
- Department of Hepatobiliary Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, China
- Organ Transplant Department, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Yongjun Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Na Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| |
Collapse
|
32
|
Nautiyal N, Maheshwari D, Kumar D, Rao EP, Tripathi DM, Kumar S, Diwakar S, Bhardwaj M, Mohanty S, Baligar P, Kumari A, Bihari C, Biswas S, Sarin SK, Kumar A. Rejuvenating bone marrow hematopoietic reserve prevents regeneration failure and hepatic decompensation in animal model of cirrhosis. Front Immunol 2024; 15:1439510. [PMID: 39188716 PMCID: PMC11345600 DOI: 10.3389/fimmu.2024.1439510] [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: 05/28/2024] [Accepted: 07/24/2024] [Indexed: 08/28/2024] Open
Abstract
Background and aim Bone marrow stem cells (BM-SCs) and their progeny play a central role in tissue repair and regeneration. In patients with chronic liver failure, bone marrow (BM) reserve is severally compromised and they showed marked defects in the resolution of injury and infection, leading to liver failure and the onset of decompensation. Whether BM failure is the cause or consequence of liver failure during cirrhosis is not known. In this study, we aimed to determine the underlying relationship between BM failure and regeneration failure in cirrhosis. Methodology C57Bl/6(J) mice were used to develop chronic liver injury through intra-peritoneal administration of carbon tetrachloride (CCl4) for 15 weeks (0.1-0.5 ml/kg). Animals were sacrificed to study the transition of cirrhosis and BM defects. To restore the BM-SC reserve; healthy BM cells were infused via intra-BM infusion and assessed for changes in liver injury, regeneration, and BM-SC reserve. Results Using a CCl4-induced animal - model of cirrhosis, we showed the loss of BM-SCs reserve occurred before regeneration failure and the onset of non-acute decompensation. Intra-BM infusion of healthy BM cells induced the repopulation of native hematopoietic stem cells (HSCs) in cirrhotic BM. Restoring BM-HSCs reserve augments liver macrophage-mediated clearance of infection and inflammation dampens neutrophil-mediated inflammation, accelerates fibrosis regression, enhances hepatocyte proliferation, and delays the onset of non-acute decompensation. Conclusion These findings suggest that loss of BM-HSCs reserve underlies the compromised innate immune function of the liver, drives regeneration failure, and the onset of non-acute decompensation. We further provide the proof-of-concept that rejuvenating BM-HSC reserve can serve as a potential therapeutic approach for preventing regeneration failure and transition to decompensated cirrhosis.
Collapse
Affiliation(s)
- Nidhi Nautiyal
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, India
| | - Deepanshu Maheshwari
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Dhananjay Kumar
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - E. Pranshu Rao
- Stem Cell Facility, All India Institute of Medical Sciences, New Delhi, India
| | - Dinesh Mani Tripathi
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Sandeep Kumar
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Sunidhi Diwakar
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Manisha Bhardwaj
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Sujata Mohanty
- Stem Cell Facility, All India Institute of Medical Sciences, New Delhi, India
| | - Prakash Baligar
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, India
| | - Anupama Kumari
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Chhagan Bihari
- Department of Pathology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Subhrajit Biswas
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, India
| | - S. K. Sarin
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Anupam Kumar
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| |
Collapse
|
33
|
Chen C, Feng D, Wang Y, Yao T, Mackowiak B, Gao B. Necrotic Liver Lesion Resolution: Another Mode of Liver Regeneration. Semin Liver Dis 2024; 44:333-342. [PMID: 38955211 DOI: 10.1055/a-2358-9505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
The liver has the great ability to regenerate after partial resection or injury, and the mechanisms underlying liver regeneration have been extensively investigated. Interestingly, acute liver injuries triggered by various etiologies are associated with the formation of necrotic lesions, and such necrotic lesions are also rapidly resolved. However, how necrotic liver lesions are repaired has not been carefully investigated until recently. In this review, we briefly summarize the spatiotemporal process of necrotic liver lesion resolution in several liver injury models including immune-mediated liver injury and drug-induced liver injury. The roles of liver nonparenchymal cells and infiltrating immune cells in controlling necrotic liver lesion resolution are discussed, which may help identify potential therapies for acute liver injury and failure.
Collapse
Affiliation(s)
- Cheng Chen
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Dechun Feng
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Yang Wang
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Tiantian Yao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Bryan Mackowiak
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
34
|
Yan L, Wang J, Cai X, Liou Y, Shen H, Hao J, Huang C, Luo G, He W. Macrophage plasticity: signaling pathways, tissue repair, and regeneration. MedComm (Beijing) 2024; 5:e658. [PMID: 39092292 PMCID: PMC11292402 DOI: 10.1002/mco2.658] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 08/04/2024] Open
Abstract
Macrophages are versatile immune cells with remarkable plasticity, enabling them to adapt to diverse tissue microenvironments and perform various functions. Traditionally categorized into classically activated (M1) and alternatively activated (M2) phenotypes, recent advances have revealed a spectrum of macrophage activation states that extend beyond this dichotomy. The complex interplay of signaling pathways, transcriptional regulators, and epigenetic modifications orchestrates macrophage polarization, allowing them to respond to various stimuli dynamically. Here, we provide a comprehensive overview of the signaling cascades governing macrophage plasticity, focusing on the roles of Toll-like receptors, signal transducer and activator of transcription proteins, nuclear receptors, and microRNAs. We also discuss the emerging concepts of macrophage metabolic reprogramming and trained immunity, contributing to their functional adaptability. Macrophage plasticity plays a pivotal role in tissue repair and regeneration, with macrophages coordinating inflammation, angiogenesis, and matrix remodeling to restore tissue homeostasis. By harnessing the potential of macrophage plasticity, novel therapeutic strategies targeting macrophage polarization could be developed for various diseases, including chronic wounds, fibrotic disorders, and inflammatory conditions. Ultimately, a deeper understanding of the molecular mechanisms underpinning macrophage plasticity will pave the way for innovative regenerative medicine and tissue engineering approaches.
Collapse
Affiliation(s)
- Lingfeng Yan
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| | - Jue Wang
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| | - Xin Cai
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| | - Yih‐Cherng Liou
- Department of Biological SciencesFaculty of ScienceNational University of SingaporeSingaporeSingapore
- National University of Singapore (NUS) Graduate School for Integrative Sciences and EngineeringNational University of SingaporeSingaporeSingapore
| | - Han‐Ming Shen
- Faculty of Health SciencesUniversity of MacauMacauChina
| | - Jianlei Hao
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and TreatmentZhuhai Institute of Translational MedicineZhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University)Jinan UniversityZhuhaiGuangdongChina
- The Biomedical Translational Research InstituteFaculty of Medical ScienceJinan UniversityGuangzhouGuangdongChina
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospitaland West China School of Basic Medical Sciences and Forensic MedicineSichuan University, and Collaborative Innovation Center for BiotherapyChengduChina
| | - Gaoxing Luo
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| | - Weifeng He
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| |
Collapse
|
35
|
Wei H, Yi K, Li F, Li D, Yang J, Shi R, Jin Y, Wang H, Ding J, Tao Y, Li M. Multimodal Tetrahedral DNA Nanoplatform for Surprisingly Rapid and Significant Treatment of Acute Liver Failure. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2305826. [PMID: 37801371 DOI: 10.1002/adma.202305826] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 09/07/2023] [Indexed: 10/08/2023]
Abstract
Acute liver failure (ALF) is a life-threatening disease associated with the rapid development of inflammatory storms, level elevation of reactive oxygen species (ROS), and hepatocyte necrosis, which results in high short-term mortality. Except for liver transplantation, no effective strategies are available for ALF therapy due to the rapid disease progression and narrow window of therapeutic time. Therefore, there is an urgent demand to explore the fast and effective modalities for ALF treatment. Herein, a multifunctional tetrahedral DNA nanoplatform (TDN) is constructed by incorporating tumor necrosis factor-α siRNA (siTNF-α) through DNA hybridization and antioxidant manganese porphyrin (MnP4) via π-π stacking interaction with G-quadruplex (G4) for surprisingly rapid and significant ALF therapy. TDN-siTNF-α/-G4-MnP4 silences TNF-α of macrophages by siTNF-α and polarizes them to the anti-inflammatory M2 phenotype, providing appropriate microenvironments for hepatocyte viability. Additionally, TDN-siTNF-α/-G4-MnP4 scavenges intracellular ROS by MnP4, protecting hepatocytes from oxidative-stress-associated cell death. Furthermore, TDN itself promotes hepatocyte proliferation by modulating the cell cycle. TDN-siTNF-α/-G4-MnP4 shows almost complete liver accumulation after intravenous injection and exhibits excellent therapeutic efficacy of ALF within 2 h. The multifunctional DNA nanoformulation provides an effective strategy for rapid ALF therapy, expanding its application for innovative treatments of liver diseases.
Collapse
Affiliation(s)
- Hongyan Wei
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, P. R. China
- Guangdong Provincial Key Laboratory of Liver Disease, 600 Tianhe Road, Guangzhou, 510630, P. R. China
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children, 120 Longshan Road, Chongqing, 401147, P. R. China
| | - Ke Yi
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, P. R. China
| | - Fenfang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, P. R. China
| | - Di Li
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
| | - Jiazhen Yang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
| | - Run Shi
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, P. R. China
| | - Yuanyuan Jin
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, P. R. China
| | - Haixia Wang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, 220 Handan Road, Shanghai, 200433, P. R. China
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, P. R. China
- Guangdong Provincial Key Laboratory of Liver Disease, 600 Tianhe Road, Guangzhou, 510630, P. R. China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, P. R. China
- Guangdong Provincial Key Laboratory of Liver Disease, 600 Tianhe Road, Guangzhou, 510630, P. R. China
| |
Collapse
|
36
|
Dai H, Zhu C, Huai Q, Xu W, Zhu J, Zhang X, Zhang X, Sun B, Xu H, Zheng M, Li X, Wang H. Chimeric antigen receptor-modified macrophages ameliorate liver fibrosis in preclinical models. J Hepatol 2024; 80:913-927. [PMID: 38340812 DOI: 10.1016/j.jhep.2024.01.034] [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: 06/21/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND & AIMS Treatments directly targeting fibrosis remain limited. Given the unique intrinsic features of macrophages and their capacity to engraft in the liver, we genetically engineered bone marrow-derived macrophages with a chimeric antigen receptor (CAR) to direct their phagocytic activity against hepatic stellate cells (HSCs) in multiple mouse models. This study aimed to demonstrate the therapeutic efficacy of CAR macrophages (CAR-Ms) in mouse models of fibrosis and cirrhosis and to elucidate the underlying mechanisms. METHODS uPAR expression was studied in patients with fibrosis/cirrhosis and in murine models of liver fibrosis, including mice treated with carbon tetrachloride, a 5-diethoxycarbonyl-1, 4-dihydrocollidine diet, or a high-fat/cholesterol/fructose diet. The safety and efficacy of CAR-Ms were evaluated in vitro and in vivo. RESULTS Adoptive transfer of CAR-Ms resulted in a significant reduction in liver fibrosis and the restoration of function in murine models of liver fibrosis. CAR-Ms modulated the hepatic immune microenvironment to recruit and modify the activation of endogenous immune cells to drive fibrosis regression. These CAR-Ms were able to recruit and present antigens to T cells and mount specific antifibrotic T-cell responses to reduce fibroblasts and liver fibrosis in mice. CONCLUSION Collectively, our findings demonstrate the potential of using macrophages as a platform for CAR technology to provide an effective treatment option for liver fibrosis. CAR-Ms might be developed for treatment of patients with liver fibrosis. IMPACT AND IMPLICATIONS Liver fibrosis is an incurable condition that afflicts millions of people globally. Despite the clear clinical need, therapies for liver fibrosis are limited. Our findings provide the first preclinical evidence that chimeric antigen receptor (CAR)-macrophages (CAR-Ms) targeting uPAR can attenuate liver fibrosis and cirrhosis. We show that macrophages expressing this uPAR CAR exert a direct antifibrotic effect and elicit a specific T-cell response that augments the immune response against liver fibrosis. These findings demonstrate the potential of using CAR-Ms as an effective cell-based therapy for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Hanren Dai
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Cheng Zhu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Qian Huai
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Wentao Xu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Jiejie Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xu Zhang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Xianzheng Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Beicheng Sun
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Honghai Xu
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Minghua Zheng
- MAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaolei Li
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China; Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China.
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China.
| |
Collapse
|
37
|
Ashmore-Harris C, Antonopoulou E, Finney SM, Vieira MR, Hennessy MG, Muench A, Lu WY, Gadd VL, El Haj AJ, Forbes SJ, Waters SL. Exploiting in silico modelling to enhance translation of liver cell therapies from bench to bedside. NPJ Regen Med 2024; 9:19. [PMID: 38724586 PMCID: PMC11081951 DOI: 10.1038/s41536-024-00361-3] [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: 07/05/2023] [Accepted: 04/18/2024] [Indexed: 05/12/2024] Open
Abstract
Cell therapies are emerging as promising treatments for a range of liver diseases but translational bottlenecks still remain including: securing and assessing the safe and effective delivery of cells to the disease site; ensuring successful cell engraftment and function; and preventing immunogenic responses. Here we highlight three therapies, each utilising a different cell type, at different stages in their clinical translation journey: transplantation of multipotent mesenchymal stromal/signalling cells, hepatocytes and macrophages. To overcome bottlenecks impeding clinical progression, we advocate for wider use of mechanistic in silico modelling approaches. We discuss how in silico approaches, alongside complementary experimental approaches, can enhance our understanding of the mechanisms underlying successful cell delivery and engraftment. Furthermore, such combined theoretical-experimental approaches can be exploited to develop novel therapies, address safety and efficacy challenges, bridge the gap between in vitro and in vivo model systems, and compensate for the inherent differences between animal model systems and humans. We also highlight how in silico model development can result in fewer and more targeted in vivo experiments, thereby reducing preclinical costs and experimental animal numbers and potentially accelerating translation to the clinic. The development of biologically-accurate in silico models that capture the mechanisms underpinning the behaviour of these complex systems must be reinforced by quantitative methods to assess cell survival post-transplant, and we argue that non-invasive in vivo imaging strategies should be routinely integrated into transplant studies.
Collapse
Affiliation(s)
- Candice Ashmore-Harris
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | | | - Simon M Finney
- Mathematical Institute, University of Oxford, Oxford, OX2 6GG, UK
| | - Melissa R Vieira
- Healthcare Technologies Institute (HTI), Institute of Translational Medicine, University of Birmingham, Birmingham, B15 2TH, UK
- School of Chemical Engineering, College of Engineering and Physical Sciences, University of Birmingham, Birmingham, B15 2TH, UK
| | - Matthew G Hennessy
- Department of Engineering Mathematics, University of Bristol, BS8 1TW, Bristol, UK
| | - Andreas Muench
- Mathematical Institute, University of Oxford, Oxford, OX2 6GG, UK
| | - Wei-Yu Lu
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Victoria L Gadd
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Alicia J El Haj
- Healthcare Technologies Institute (HTI), Institute of Translational Medicine, University of Birmingham, Birmingham, B15 2TH, UK
- School of Chemical Engineering, College of Engineering and Physical Sciences, University of Birmingham, Birmingham, B15 2TH, UK
| | - Stuart J Forbes
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Sarah L Waters
- Mathematical Institute, University of Oxford, Oxford, OX2 6GG, UK.
| |
Collapse
|
38
|
Camps G, Maestro S, Torella L, Herrero D, Usai C, Bilbao-Arribas M, Aldaz A, Olagüe C, Vales A, Suárez-Amarán L, Aldabe R, Gonzalez-Aseguinolaza G. Protective role of RIPK1 scaffolding against HDV-induced hepatocyte cell death and the significance of cytokines in mice. PLoS Pathog 2024; 20:e1011749. [PMID: 38739648 PMCID: PMC11115361 DOI: 10.1371/journal.ppat.1011749] [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: 10/15/2023] [Revised: 05/23/2024] [Accepted: 04/16/2024] [Indexed: 05/16/2024] Open
Abstract
Hepatitis delta virus (HDV) infection represents the most severe form of human viral hepatitis; however, the mechanisms underlying its pathology remain incompletely understood. We recently developed an HDV mouse model by injecting adeno-associated viral vectors (AAV) containing replication-competent HBV and HDV genomes. This model replicates many features of human infection, including liver injury. Notably, the extent of liver damage can be diminished with anti-TNF-α treatment. Here, we found that TNF-α is mainly produced by macrophages. Downstream of the TNF-α receptor (TNFR), the receptor-interacting serine/threonine-protein kinase 1 (RIPK1) serves as a cell fate regulator, playing roles in both cell survival and death pathways. In this study, we explored the function of RIPK1 and other host factors in HDV-induced cell death. We determined that the scaffolding function of RIPK1, and not its kinase activity, offers partial protection against HDV-induced apoptosis. A reduction in RIPK1 expression in hepatocytes through CRISPR-Cas9-mediated gene editing significantly intensifies HDV-induced damage. Contrary to our expectations, the protective effect of RIPK1 was not linked to TNF-α or macrophage activation, as their absence did not alter the extent of damage. Intriguingly, in the absence of RIPK1, macrophages confer a protective role. However, in animals unresponsive to type-I IFNs, RIPK1 downregulation did not exacerbate the damage, suggesting RIPK1's role in shielding hepatocytes from type-I IFN-induced cell death. Interestingly, while the damage extent is similar between IFNα/βR KO and wild type mice in terms of transaminase elevation, their cell death mechanisms differ. In conclusion, our findings reveal that HDV-induced type-I IFN production is central to inducing hepatocyte death, and RIPK1's scaffolding function offers protective benefits. Thus, type-I IFN together with TNF-α, contribute to HDV-induced liver damage. These insights may guide the development of novel therapeutic strategies to mitigate HDV-induced liver damage and halt disease progression.
Collapse
Affiliation(s)
- Gracián Camps
- DNA & RNA Medicine Division, CIMA, University of Navarra, Instituto de Investigación Sanitaria de Navarra, IdisNA, Pamplona, Spain
| | - Sheila Maestro
- DNA & RNA Medicine Division, CIMA, University of Navarra, Instituto de Investigación Sanitaria de Navarra, IdisNA, Pamplona, Spain
| | - Laura Torella
- DNA & RNA Medicine Division, CIMA, University of Navarra, Instituto de Investigación Sanitaria de Navarra, IdisNA, Pamplona, Spain
| | - Diego Herrero
- DNA & RNA Medicine Division, CIMA, University of Navarra, Instituto de Investigación Sanitaria de Navarra, IdisNA, Pamplona, Spain
| | - Carla Usai
- DNA & RNA Medicine Division, CIMA, University of Navarra, Instituto de Investigación Sanitaria de Navarra, IdisNA, Pamplona, Spain
| | - Martin Bilbao-Arribas
- DNA & RNA Medicine Division, CIMA, University of Navarra, Instituto de Investigación Sanitaria de Navarra, IdisNA, Pamplona, Spain
| | - Ana Aldaz
- DNA & RNA Medicine Division, CIMA, University of Navarra, Instituto de Investigación Sanitaria de Navarra, IdisNA, Pamplona, Spain
| | - Cristina Olagüe
- DNA & RNA Medicine Division, CIMA, University of Navarra, Instituto de Investigación Sanitaria de Navarra, IdisNA, Pamplona, Spain
| | - Africa Vales
- DNA & RNA Medicine Division, CIMA, University of Navarra, Instituto de Investigación Sanitaria de Navarra, IdisNA, Pamplona, Spain
| | - Lester Suárez-Amarán
- DNA & RNA Medicine Division, CIMA, University of Navarra, Instituto de Investigación Sanitaria de Navarra, IdisNA, Pamplona, Spain
| | - Rafael Aldabe
- DNA & RNA Medicine Division, CIMA, University of Navarra, Instituto de Investigación Sanitaria de Navarra, IdisNA, Pamplona, Spain
| | - Gloria Gonzalez-Aseguinolaza
- DNA & RNA Medicine Division, CIMA, University of Navarra, Instituto de Investigación Sanitaria de Navarra, IdisNA, Pamplona, Spain
| |
Collapse
|
39
|
Wang Y, Rodrigues RM, Chen C, Feng D, Maccioni L, Gao B. Macrophages in necrotic liver lesion repair: opportunities for therapeutical applications. Am J Physiol Cell Physiol 2024; 326:C1556-C1562. [PMID: 38618702 PMCID: PMC11371317 DOI: 10.1152/ajpcell.00053.2024] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/25/2024] [Accepted: 03/25/2024] [Indexed: 04/16/2024]
Abstract
Healthy livers contain 80% of body resident macrophages known as Kupffer cells. In diseased livers, the number of Kupffer cells usually drops but is compensated by infiltration of monocyte-derived macrophages, some of which can differentiate into Kupffer-like cells. Early studies suggest that Kupffer cells play important roles in both promoting liver injury and liver regeneration. Yet, the distinction between the functionalities of resident and infiltrating macrophages is not always made. By using more specific macrophage markers and targeted cell depletion and single-cell RNA sequencing, recent studies revealed several subsets of monocyte-derived macrophages that play important functions in inducing liver damage and inflammation as well as in liver repair and regeneration. In this review, we discuss the different roles that hepatic macrophages play in promoting necrotic liver lesion resolution and dead cell clearance, as well as the targeting of these cells as potential tools for the development of novel therapies for acute liver failure and acute-on-chronic liver failure.
Collapse
Affiliation(s)
- Yang Wang
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, United States
| | - Robim M Rodrigues
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, B1090, Brussels, Belgium
| | - Cheng Chen
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, United States
| | - Dechun Feng
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, United States
| | - Luca Maccioni
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, United States
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, United States
| |
Collapse
|
40
|
Hu W, Zhang X, Liu Z, Yang J, Sheng H, Liu Z, Chen C, Shang R, Chen Y, Lu Y, Hu X, Huang Y, Yin W, Cai X, Fan D, Yan L, Hao J, Luo G, He W. Spatiotemporal orchestration of macrophage activation trajectories by Vγ4 T cells during skin wound healing. iScience 2024; 27:109545. [PMID: 38617557 PMCID: PMC11015460 DOI: 10.1016/j.isci.2024.109545] [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: 10/25/2023] [Revised: 02/08/2024] [Accepted: 03/18/2024] [Indexed: 04/16/2024] Open
Abstract
Dysregulated macrophage polarization from pro-inflammatory M1 to anti-inflammatory M2 phenotypes underlies impaired cutaneous wound healing. This study reveals Vγ4+ γδ T cells spatiotemporally calibrate macrophage trajectories during skin repair via sophisticated interferon-γ (IFN-γ) conditioning across multiple interconnected tissues. Locally within wound beds, infiltrating Vγ4+ γδ T cells directly potentiate M1 activation and suppress M2 polarization thereby prolonging local inflammation. In draining lymph nodes, infiltrated Vγ4+ γδ T cells expand populations of IFN-γ-competent lymphocytes which disseminate systemically and infiltrate into wound tissues, further enforcing M1 macrophages programming. Moreover, Vγ4+γδ T cells flushed into bone marrow stimulate increased IFN-γ production, which elevates the output of pro-inflammatory Ly6C+monocytes. Mobilization of these monocytes continually replenishes the M1 macrophage pool in wounds, preventing phenotypic conversion to M2 activation. Thus, multi-axis coordination of macrophage activation trajectories by trafficking Vγ4+ γδ T cells provides a sophisticated immunological mechanism regulating inflammation timing and resolution during skin repair.
Collapse
Affiliation(s)
- Wengang Hu
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing 400038, China
| | - Xiaorong Zhang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing 400038, China
| | - Zhongyang Liu
- Department of Plastic Surgery, the First Affiliated Hospital, Zhengzhou University, Henan, China
| | - Jiacai Yang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing 400038, China
| | - Hao Sheng
- Urology Department, the Second Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Zhihui Liu
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing 400038, China
| | - Cheng Chen
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing 400038, China
| | - Ruoyu Shang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing 400038, China
| | - Yunxia Chen
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing 400038, China
| | - Yifei Lu
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing 400038, China
| | - Xiaohong Hu
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing 400038, China
| | - Yong Huang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing 400038, China
| | - Wenjing Yin
- Academy of Biological Engineering, Chongqing University, Chongqing, China
| | - Xin Cai
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing 400038, China
| | - Dejiang Fan
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing 400038, China
| | - Lingfeng Yan
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing 400038, China
| | - Jianlei Hao
- Zhuhai Institute of Translational Medicine, Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai 519000 Guangdong, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou 510632, Guangdong, China
| | - Gaoxing Luo
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing 400038, China
| | - Weifeng He
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing 400038, China
| |
Collapse
|
41
|
Feng L, Wang Y, Fu Y, Li T, He G. Stem Cell-Based Strategies: The Future Direction of Bioartificial Liver Development. Stem Cell Rev Rep 2024; 20:601-616. [PMID: 38170319 DOI: 10.1007/s12015-023-10672-5] [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] [Accepted: 12/21/2023] [Indexed: 01/05/2024]
Abstract
Acute liver failure (ALF) results from severe liver damage or end-stage liver disease. It is extremely fatal and causes serious health and economic burdens worldwide. Once ALF occurs, liver transplantation (LT) is the only definitive and recommended treatment; however, LT is limited by the scarcity of liver grafts. Consequently, the clinical use of bioartificial liver (BAL) has been proposed as a treatment strategy for ALF. Human primary hepatocytes are an ideal cell source for these methods. However, their high demand and superior viability prevent their widespread use. Hence, finding alternatives that meet the seed cell quality and quantity requirements is imperative. Stem cells with self-renewing, immunogenic, and differentiative capacities are potential cell sources. MSCs and its secretomes encompass a spectrum of beneficial properties, such as anti-inflammatory, immunomodulatory, anti-ROS (reactive oxygen species), anti-apoptotic, pro-metabolomic, anti-fibrogenesis, and pro-regenerative attributes. This review focused on the recent status and future directions of stem cell-based strategies in BAL for ALF. Additionally, we discussed the opportunities and challenges associated with promoting such strategies for clinical applications.
Collapse
Affiliation(s)
- Lei Feng
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China.
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550000, Guizhou, China.
| | - Yi Wang
- Shanxi Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, Shanxi, China
| | - Yu Fu
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Ting Li
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China.
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510140, Guangdong, China.
| | - Guolin He
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China.
| |
Collapse
|
42
|
Chen Y, Wang J, Zhou N, Fang Q, Cai H, Du Z, An R, Liu D, Chen X, Wang X, Li F, Yan Q, Chen L, Du J. Protozoan-Derived Cytokine-Transgenic Macrophages Reverse Hepatic Fibrosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308750. [PMID: 38247166 PMCID: PMC10987136 DOI: 10.1002/advs.202308750] [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: 11/15/2023] [Revised: 01/01/2024] [Indexed: 01/23/2024]
Abstract
Macrophage therapy for liver fibrosis is on the cusp of meaningful clinical utility. Due to the heterogeneities of macrophages, it is urgent to develop safer macrophages with a more stable and defined phenotype for the treatment of liver fibrosis. Herein, a new macrophage-based immunotherapy using macrophages stably expressing a pivotal cytokine from Toxoplasma gondii, a parasite that infects ≈ 2 billion people is developed. It is found that Toxoplasma gondii macrophage migration inhibitory factor-transgenic macrophage (Mφtgmif) shows stable fibrinolysis and strong chemotactic capacity. Mφtgmif effectively ameliorates liver fibrosis and deactivates aHSCs by recruiting Ly6Chi macrophages via paracrine CCL2 and polarizing them into the restorative Ly6Clo macrophage through the secretion of CX3CL1. Remarkably, Mφtgmif exhibits even higher chemotactic potential, lower grade of inflammation, and better therapeutic effects than LPS/IFN-γ-treated macrophages, making macrophage-based immune therapy more efficient and safer. Mechanistically, TgMIF promotes CCL2 expression by activating the ERK/HMGB1/NF-κB pathway, and this event is associated with recruiting endogenous macrophages into the fibrosis liver. The findings do not merely identify viable immunotherapy for liver fibrosis but also suggest a therapeutic strategy based on the evolutionarily designed immunomodulator to treat human diseases by modifying the immune microenvironment.
Collapse
Affiliation(s)
- Ying Chen
- Department of Biochemistry and Molecular BiologyResearch Center for Infectious DiseasesSchool of Basic Medical SciencesAnhui Medical UniversityHefei230032China
- The Provincial Key Laboratory of Zoonoses of High Institutions in AnhuiAnhui Medical UniversityHefei230032China
- School of NursingAnhui Medical UniversityHefei230032China
| | - Jie Wang
- Department of Biochemistry and Molecular BiologyResearch Center for Infectious DiseasesSchool of Basic Medical SciencesAnhui Medical UniversityHefei230032China
- The Provincial Key Laboratory of Zoonoses of High Institutions in AnhuiAnhui Medical UniversityHefei230032China
| | - Nan Zhou
- Department of Biochemistry and Molecular BiologyResearch Center for Infectious DiseasesSchool of Basic Medical SciencesAnhui Medical UniversityHefei230032China
- The Provincial Key Laboratory of Zoonoses of High Institutions in AnhuiAnhui Medical UniversityHefei230032China
| | - Qi Fang
- Department of AnesthesiologyThe First Affiliated Hospital of Anhui Medical UniversityHefei230032China
| | - Haijian Cai
- Department of Biochemistry and Molecular BiologyResearch Center for Infectious DiseasesSchool of Basic Medical SciencesAnhui Medical UniversityHefei230032China
- The Provincial Key Laboratory of Zoonoses of High Institutions in AnhuiAnhui Medical UniversityHefei230032China
| | - Zhuoran Du
- Department of Clinical MedicineWannan Medical CollegeWuhu241002China
| | - Ran An
- Department of Biochemistry and Molecular BiologyResearch Center for Infectious DiseasesSchool of Basic Medical SciencesAnhui Medical UniversityHefei230032China
- The Provincial Key Laboratory of Zoonoses of High Institutions in AnhuiAnhui Medical UniversityHefei230032China
| | - Deng Liu
- Department of AnesthesiologyThe First Affiliated Hospital of Anhui Medical UniversityHefei230032China
| | - Xuepeng Chen
- GMU‐GIBH Joint School of Life SciencesThe Guangdong‐Hong Kong‐Macau Joint Laboratory for Cell Fate Regulation and DiseasesGuangzhou National LaboratoryGuangzhou Medical UniversityGuangzhou510005China
| | - Xinxin Wang
- GMU‐GIBH Joint School of Life SciencesThe Guangdong‐Hong Kong‐Macau Joint Laboratory for Cell Fate Regulation and DiseasesGuangzhou National LaboratoryGuangzhou Medical UniversityGuangzhou510005China
| | - Fangmin Li
- Department of Biochemistry and Molecular BiologyResearch Center for Infectious DiseasesSchool of Basic Medical SciencesAnhui Medical UniversityHefei230032China
- The Provincial Key Laboratory of Zoonoses of High Institutions in AnhuiAnhui Medical UniversityHefei230032China
| | - Qi Yan
- Department of Biochemistry and Molecular BiologyResearch Center for Infectious DiseasesSchool of Basic Medical SciencesAnhui Medical UniversityHefei230032China
- The Provincial Key Laboratory of Zoonoses of High Institutions in AnhuiAnhui Medical UniversityHefei230032China
| | - Lijian Chen
- Department of AnesthesiologyThe First Affiliated Hospital of Anhui Medical UniversityHefei230032China
| | - Jian Du
- Department of Biochemistry and Molecular BiologyResearch Center for Infectious DiseasesSchool of Basic Medical SciencesAnhui Medical UniversityHefei230032China
- The Provincial Key Laboratory of Zoonoses of High Institutions in AnhuiAnhui Medical UniversityHefei230032China
| |
Collapse
|
43
|
Sharma S, Ghufran SM, Aftab M, Bihari C, Ghose S, Biswas S. Survivin inhibition ameliorates liver fibrosis by inducing hepatic stellate cell senescence and depleting hepatic macrophage population. J Cell Commun Signal 2024; 18:e12015. [PMID: 38545255 PMCID: PMC10964939 DOI: 10.1002/ccs3.12015] [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: 11/24/2023] [Accepted: 12/28/2023] [Indexed: 01/29/2025] Open
Abstract
Persistent activation of hepatic stellate cells (HSCs) in the injured liver leads to the progression of liver injury from fibrosis to detrimental cirrhosis. In a previous study, we have shown that survivin protein is upregulated during the early activation of HSCs, which triggers the onset of liver fibrosis. However, the therapeutic potential of targeting survivin in a fully established fibrotic liver needs to be investigated. In this study, we chemically induced hepatic fibrosis in mice using carbon tetrachloride (CCl4) for 6 weeks, which was followed by treatment with a survivin suppressant (YM155). We also evaluated survivin expression in fibrotic human liver tissues, primary HSCs, and HSC cell line by histological analysis. αSMA+ HSCs in human and mice fibrotic liver tissues showed enhanced survivin expression, whereas the hepatocytes and quiescent (qHSCs) displayed minimal expression. Alternatively, activated M2 macrophage subtype induced survivin expression in HSCs through the TGF-β-TGF-β receptor-I/II signaling. Inhibition of survivin in HSCs promoted cell cycle arrest and senescence, which eventually suppressed their activation. In vivo, YM155 treatment increased the expression of cell senescence makers in HSCs around fibrotic septa such as p53, p21, and β-galactosidase. YM155 treatment in vivo also reduced the hepatic macrophage population and inflammatory cytokine expression in the liver. In conclusion, downregulation of survivin in the fibrotic liver decreases HSC activation by inducing cellular senescence and modulating macrophage cytokine expression that collectively ameliorates liver fibrosis.
Collapse
Affiliation(s)
- Sachin Sharma
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR)Amity UniversityNoidaUttar PradeshIndia
- Department of MedicineUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Shaikh Maryam Ghufran
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR)Amity UniversityNoidaUttar PradeshIndia
- Heersink School of MedicineUniversity of AlabamaBirminghamUSA
| | - Mehreen Aftab
- Division of Cellular and Molecular OncologyNational Institute of Cancer Prevention and Research (NICPR)NoidaUttar PradeshIndia
| | - Chhagan Bihari
- Department of PathologyInstitute of Liver and Biliary Sciences (ILBS)New DelhiIndia
| | - Sampa Ghose
- Department of Medical OncologyAll India Institute of Medical Sciences (AIIMS)New DelhiIndia
| | - Subhrajit Biswas
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR)Amity UniversityNoidaUttar PradeshIndia
| |
Collapse
|
44
|
Cui Q, Jiang T, Xie X, Wang H, Qian L, Cheng Y, Li Q, Lu T, Yao Q, Liu J, Lai B, Chen C, Xiao L, Wang N. S-nitrosylation attenuates pregnane X receptor hyperactivity and acetaminophen-induced liver injury. JCI Insight 2024; 9:e172632. [PMID: 38032737 PMCID: PMC10906221 DOI: 10.1172/jci.insight.172632] [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: 05/31/2023] [Accepted: 11/28/2023] [Indexed: 12/02/2023] Open
Abstract
Drug-induced liver injury (DILI), especially acetaminophen overdose, is the leading cause of acute liver failure. Pregnane X receptor (PXR) is a nuclear receptor and the master regulator of drug metabolism. Aberrant activation of PXR plays a pathogenic role in the acetaminophen hepatotoxicity. Here, we aimed to examine the S-nitrosylation of PXR (SNO-PXR) in response to acetaminophen. We found that PXR was S-nitrosylated in hepatocytes and the mouse livers after exposure to acetaminophen or S-nitrosoglutathione (GSNO). Mass spectrometry and site-directed mutagenesis identified the cysteine 307 as the primary residue for S-nitrosylation (SNO) modification. In hepatocytes, SNO suppressed both agonist-induced (rifampicin and SR12813) and constitutively active PXR (VP-PXR, a human PXR fused to the minimal transactivator domain of the herpes virus transcription factor VP16) activations. Furthermore, in acetaminophen-overdosed mouse livers, PXR protein was decreased at the centrilobular regions overlapping with increased SNO. In PXR-/- mice, replenishing the livers with the SNO-deficient PXR significantly aggravated hepatic necrosis, increased HMGB1 release, and exacerbated liver injury and inflammation. Particularly, we demonstrated that S-nitrosoglutathione reductase (GSNOR) inhibitor N6022 promoted hepatoprotection by increasing the levels of SNO-PXR. In conclusion, PXR is posttranslationally modified by SNO in hepatocytes in response to acetaminophen. This modification mitigated the acetaminophen-induced PXR hyperactivity. It may serve as a target for therapeutical intervention.
Collapse
Affiliation(s)
- Qi Cui
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
- School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Tingting Jiang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
- School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Xinya Xie
- School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Haodong Wang
- East China Normal University Health Science Center, Shanghai, China
| | - Lei Qian
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Yanyan Cheng
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Qiang Li
- School of Public Health, Xi’an Jiaotong University, Xi’an, China
| | - Tingxu Lu
- School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Qinyu Yao
- School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Jia Liu
- School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Baochang Lai
- School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Chang Chen
- National Laboratory of Biomacromolecules, Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Lei Xiao
- School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Nanping Wang
- East China Normal University Health Science Center, Shanghai, China
| |
Collapse
|
45
|
Kwon A, Lee NY, Yu JH, Choi MG, Park J, Koo JH. Mitochondrial stress activates YAP/TAZ through RhoA oxidation to promote liver injury. Cell Death Dis 2024; 15:51. [PMID: 38225223 PMCID: PMC10789791 DOI: 10.1038/s41419-024-06448-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 01/17/2024]
Abstract
Yes-associated protein (YAP) and WW domain-containing transcription regulator protein 1 (WWTR1; also known as TAZ) are the main effectors of the Hippo pathway and their dysregulation contributes to diseases in tissues including the liver. Although mitochondria are capable of transmitting signals to change transcriptomic landscape of diseased hepatocytes, such retrograde signaling and the related nuclear machinery are largely unknown. Here, we show that increased YAP activity is associated with mitochondrial stress during liver injury; and this is required for secondary inflammation, promoting hepatocyte death. Mitochondrial stress inducers robustly promoted YAP/TAZ dephosphorylation, nuclear accumulation, and target gene transcription. RNA sequencing revealed that the majority of mitochondrial stress transcripts required YAP/TAZ. Mechanistically, direct oxidation of RhoA by mitochondrial superoxide was responsible for PP2A-mediated YAP/TAZ dephosphorylation providing a novel physiological input for the Hippo pathway. Hepatocyte-specific Yap/Taz ablation suppressed acetaminophen-induced liver injury and blunted transcriptomic changes associated with the pathology. Our observations uncover unappreciated pathway of mitochondrial stress signaling and reveal YAP/TAZ activation as the mechanistic basis for liver injury progression.
Collapse
Affiliation(s)
- Ari Kwon
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Korea
| | - Na Young Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Korea
| | - Jae-Hyun Yu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Korea
| | - Myeung Gi Choi
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Korea
| | - Jeongwoo Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Korea
| | - Ja Hyun Koo
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Korea.
| |
Collapse
|
46
|
Zhu L, Wang Q, Guo M, Fang H, Li T, Zhu Y, Jiang H, Xiao P, Hu M. Mesenchymal Stem Cell-Derived Exosomes in Various Chronic Liver Diseases: Hype or Hope? J Inflamm Res 2024; 17:171-189. [PMID: 38223423 PMCID: PMC10788055 DOI: 10.2147/jir.s439974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 12/27/2023] [Indexed: 01/16/2024] Open
Abstract
Chronic liver conditions are associated with high mortality rates and have a large adverse effect on human well-being as well as a significant financial burden. Currently, the only effective treatment available for the effects of liver failure and cirrhosis resulting from the progression of several chronic liver diseases is liver transplantation carried out at the original location. This implies that developing novel and effective treatments is imperative. Regenerative medicine has long been associated with stem cell therapy. Mesenchymal stem cells (MSCs), a type of cell with great differentiation potential, have become the preferred source for stem cell therapy. According to recent studies, MSCs' paracrine products-rather than their capacity for differentiation-play a significant therapeutic effect. MSC exosomes, a type of extracellular vesicle (MSC-EV), came into view as the paracrine substances of MSCs. According to research, MSC exosomes can maintain tissue homeostasis, which is necessary for healthy tissue function. All tissues contain them, and they take part in a variety of biological activities that support cellular activity and tissue regeneration in order to preserve tissue homeostasis. The outcomes support the use of MSCs and the exosomes they produce as a therapeutic option for a range of diseases. This review provides a brief overview of the source of MSC-EVs and outlines their physiological roles and biochemical capabilities. The elucidation of the role of MSC-EVs in the recovery and repair of hepatic tissues, as well as their contribution to maintaining tissue homeostasis, is discussed in relation to different chronic liver diseases. This review aims to provide new insights into the unique roles that MSC-EVs play in the treatment of chronic liver diseases.
Collapse
Affiliation(s)
- Lujian Zhu
- Department of Infectious Diseases, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People’s Republic of China
| | - Qin Wang
- Department of Infectious Diseases, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People’s Republic of China
| | - Maodong Guo
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People’s Republic of China
| | - Hao Fang
- Department of Traumatology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People’s Republic of China
| | - Ting Li
- Department of Emergency Medicine, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Yin Zhu
- Department of Infectious Diseases, Taizhou Enze Medical Center (Group), Enze Hospital, Taizhou, People’s Republic of China
| | - Huimian Jiang
- Department of Infectious Diseases, the First Affiliated Hospital of Ningbo University, Ningbo, People’s Republic of China
| | - Peiguang Xiao
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People’s Republic of China
| | - Minli Hu
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People’s Republic of China
| |
Collapse
|
47
|
Puengel T, Tacke F. Role of Kupffer cells and other immune cells. SINUSOIDAL CELLS IN LIVER DISEASES 2024:483-511. [DOI: 10.1016/b978-0-323-95262-0.00024-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
48
|
Shi H, Moore MP, Wang X, Tabas I. Efferocytosis in liver disease. JHEP Rep 2024; 6:100960. [PMID: 38234410 PMCID: PMC10792655 DOI: 10.1016/j.jhepr.2023.100960] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/11/2023] [Accepted: 10/17/2023] [Indexed: 01/19/2024] Open
Abstract
The process of dead cell clearance by phagocytic cells, called efferocytosis, prevents inflammatory cell necrosis and promotes resolution and repair. Defective efferocytosis contributes to the progression of numerous diseases in which cell death is prominent, including liver disease. Many gaps remain in our understanding of how hepatic macrophages carry out efferocytosis and how this process goes awry in various types of liver diseases. Thus far, studies have suggested that, upon liver injury, liver-resident Kupffer cells and infiltrating monocyte-derived macrophages clear dead cells, limit inflammation, and, through macrophage reprogramming, repair liver damage. However, in unusual settings, efferocytosis can promote liver disease. In this review, we will focus on efferocytosis in various types of acute and chronic liver diseases, including metabolic dysfunction-associated steatohepatitis. Understanding the mechanisms and consequences of efferocytosis by hepatic macrophages has the potential to shed new light on liver disease pathophysiology and to guide new treatment strategies to prevent disease progression.
Collapse
Affiliation(s)
- Hongxue Shi
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Mary P. Moore
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Xiaobo Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Ira Tabas
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
49
|
Lin J, Liu Y, Liu P, Qi W, Liu J, He X, Liu Q, Liu Z, Yin J, Lin J, Bao H, Lin J. SNHG17 alters anaerobic glycolysis by resetting phosphorylation modification of PGK1 to foster pro-tumor macrophage formation in pancreatic ductal adenocarcinoma. J Exp Clin Cancer Res 2023; 42:339. [PMID: 38098044 PMCID: PMC10722693 DOI: 10.1186/s13046-023-02890-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 11/06/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Within the tumor immune microenvironment (TME), tumor-associated macrophages (TAMs) are crucial in modulating polarization states to influence cancer development through metabolic reprogramming. While long non-coding RNAs (lncRNAs) have been shown to play a pivotal role in the progression of various cancers, the underlying mechanisms by which lncRNAs alter M2 polarization through macrophage metabolism remodeling remain unelucidated. METHODS RNA sequencing was used to screen for differentially expressed lncRNAs in TAMs and normal tissue-resident macrophages (NTRMs) isolated from pancreatic ductal adenocarcinoma (PDAC) tissues, whilst RT-qPCR and FISH were employed to detect the expression level of SNHG17. Moreover, a series of in vivo and in vitro experiments were conducted to assess the functions of SNHG17 from TAMs in the polarization and glycolysis of M2-like macrophages and in the proliferation and metastasis of pancreatic cancer cells (PCs). Furthermore, Western blotting, RNA pull-down, mass spectrometry, RIP, and dual-luciferase assays were utilized to explore the underlying mechanism through which SNHG17 induces pro-tumor macrophage formation. RESULTS SNHG17 was substantially enriched in TAMs and was positively correlated with a worse prognosis in PDAC. Meanwhile, functional assays determined that SNHG17 promoted the malignant progression of PCs by enhancing M2 macrophage polarization and anaerobic glycolysis. Mechanistically, SNHG17 could sponge miR-628-5p to release PGK1 mRNA and concurrently interact with the PGK1 protein, activating the pro-tumorigenic function of PGK1 by enhancing phosphorylation at the T168A site of PGK1 through ERK1/2 recruitment. Lastly, SNHG17 knockdown could reverse the polarization status of macrophages in PDAC. CONCLUSIONS The present study illustrated the essential role of SNHG17 and its molecular mechanism in TAMs derived from PDAC, indicating that SNHG17 might be a viable target for PDAC immunotherapy.
Collapse
Affiliation(s)
- Jiayu Lin
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Yihao Liu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Pengyi Liu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Wenxin Qi
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jia Liu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Xingfeng He
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qian Liu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zehua Liu
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, 00014, Helsinki, Finland
| | - Jingxin Yin
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Jiewei Lin
- Research Institute of Pancreatic Disease, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Haili Bao
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- School of Life Sciences, Shanghai University, Shanghai, China.
| | - Jianhong Lin
- Department of Pharmacy, The Third Hospital of Xiamen, Xiamen, 361100, China.
| |
Collapse
|
50
|
Ping D, Peng Y, Hu X, Liu C. Macrophage cytotherapy on liver cirrhosis. Front Pharmacol 2023; 14:1265935. [PMID: 38161689 PMCID: PMC10757375 DOI: 10.3389/fphar.2023.1265935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 12/04/2023] [Indexed: 01/03/2024] Open
Abstract
Macrophages, an essential cell population involved in mediating innate immunity in the host, play a crucial role on the development of hepatic cirrhosis. Extensive studies have highlighted the potential therapeutic benefits of macrophage therapy in treating hepatic cirrhosis. This review aims to provide a comprehensive overview of the various effects and underlying mechanisms associated with macrophage therapy in the context of hepatic cirrhosis.
Collapse
Affiliation(s)
- Dabing Ping
- Institute of Liver Diseases, Shuguang Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuan Peng
- Institute of Liver Diseases, Shuguang Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xudong Hu
- Department of Biology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chenghai Liu
- Institute of Liver Diseases, Shuguang Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai, China
| |
Collapse
|