1
|
Zhang J, Xie Z, Zhu X, Xu C, Lin J, Zhao M, Cheng Y. New insights into therapeutic strategies for targeting hepatic macrophages to alleviate liver fibrosis. Int Immunopharmacol 2025; 158:114864. [PMID: 40378438 DOI: 10.1016/j.intimp.2025.114864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/29/2025] [Accepted: 05/09/2025] [Indexed: 05/18/2025]
Abstract
Liver fibrosis is a wound-healing response induced by persistent liver damage, resulting from complex multicellular interactions and multifactorial networks. Without intervention, it can progress to cirrhosis and even liver cancer. Current understanding suggests that liver fibrosis is reversible, making it crucial to explore effective therapeutic strategies for its alleviation. Chronic inflammation serves as the primary driver of liver fibrosis, with hepatic macrophages playing a dual role depending on their polarization state. This review summarizes various prevention and therapeutic strategies targeting hepatic macrophages in the context of liver fibrosis. These strategies include inhibition of macrophage recruitment, modulation of macrophage activation and polarization, regulation of macrophage metabolism, and induction of phagocytosis and autophagy in hepatic macrophages. Additionally, we discuss the communication between hepatic macrophages, hepatocytes, and hepatic stellate cells (HSCs), as well as the current clinical application of anti-fibrotic drugs targeting macrophages. The goal is to identify effective therapeutic targets at each stage of macrophage participation in liver fibrosis development, with the aim of using hepatic macrophages as a target for liver fibrosis treatment.
Collapse
Affiliation(s)
- Jialu Zhang
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Zhaojing Xie
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Xueyu Zhu
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Chenxi Xu
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Jiguo Lin
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Mingqi Zhao
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Yunyun Cheng
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China.
| |
Collapse
|
2
|
Jaeschke H, Ramachandran A. The multiple mechanisms and modes of cell death after acetaminophen overdose. EXPLORATION OF DIGESTIVE DISEASES 2025; 4:100569. [PMID: 40364831 PMCID: PMC12074662 DOI: 10.37349/edd.2025.100569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 03/25/2025] [Indexed: 05/15/2025]
Abstract
Acetaminophen (APAP)-induced liver injury and acute liver failure is a significant clinical problem worldwide; in addition, APAP overdoses in animals or in cell culture are used as popular models to study drug-induced liver injury mechanisms and test therapeutic interventions. Early assumptions that APAP toxicity is caused by a single mechanism resulting in a defined mode of cell death in hepatocytes had to be questioned when over the years many different mechanisms and modes of cell death were reported. Although many of the contradictory results and conclusions reported over the years can be attributed to lack of understanding of established mechanisms, methodological problems, and misinterpretation of data, it is increasingly recognized that some of the reported differences in signaling mechanisms and even a switch in the mode of cell death can be caused by variations in the experimental conditions. In this review, examples will be discussed how experimental conditions (dose, solvent, etc.), the experimental system (species, strain, and substrain in vivo, cell type, and in vitro conditions), and also adaptive responses and off-target effects of genetic manipulations and chemical interventions, can impact the mechanisms of cell death. Given that the conditions will determine the results, it is therefore of critical importance to keep in mind the translational aspect of the experiments, i.e., the conditions relevant to the human pathophysiology. Only the full appreciation of these issues will lead to reproducible and clinically relevant results that advance our understanding of all facets of the human pathophysiology and identify clinically relevant therapeutic targets.
Collapse
Affiliation(s)
- Hartmut Jaeschke
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Anup Ramachandran
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
3
|
Kim JH, Yeo IJ, Son DJ, Han SB, Yoon DY, Lee DH, Hong JT. Chitinase 3-like protein 1 deficiency ameliorates drug-induced acute liver injury by inhibition of neutrophil recruitment through lipocalin-2. Front Pharmacol 2025; 16:1548832. [PMID: 40196357 PMCID: PMC11973357 DOI: 10.3389/fphar.2025.1548832] [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: 12/20/2024] [Accepted: 03/06/2025] [Indexed: 04/09/2025] Open
Abstract
Chitinase-3-like protein 1 (Chi3l1) is a member of the mammalian Chitinase-like protein family, and several studies reported that Chi3l1 is associated with various inflammatory diseases as well as liver diseases. Acetaminophen (APAP) is usually used for antipyretic drug, but its overdose induces acute liver injury (ALI). Several studies reported that subsequent inflammatory responses of the immune system play a critical role in the severity and outcome of APAP-induced ALI. In the present study, we investigated the role of Chi3l1 and its mechanism during APAP-induced ALI using Chi3l1 knock-out (KO) mice. We explored the function of Chi3l1 using APAP-injected KO mice and sought proteins associated with Chi3l1 through biological research data program for investigating mechanism. Liver histological analysis revealed that APAP-induced ALI was attenuated in KO mice compared to wild-type (WT) mice. We observed that APAP-induced neutrophil infiltration was decreased in the liver of KO mice compared to WT mice. To investigate this mechanism, we sought proteins potentially associated with Chi3l1 by mRNA sequencing and protein correlation analysis data. We found lipocalin-2 (Lcn2) and examined Chi3l1, Lcn2, and their relationship in the APAP-induced ALI model using recombinant proteins and antibodies. Our results suggest that Chi3l1 deficiency ameliorates APAP-induced liver injury through abrogating Lcn2-mediated neutrophil infiltration in the liver.
Collapse
Affiliation(s)
- Ji Hye Kim
- Department of Biological Sciences, Research Center of Ecomimetics, Chonnam National University, Gwangju, Republic of Korea
| | - In Jun Yeo
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
- College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea
| | - Dong Ju Son
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Sang Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Do Young Yoon
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Dong Hun Lee
- Department of Biological Sciences, Research Center of Ecomimetics, Chonnam National University, Gwangju, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| |
Collapse
|
4
|
Livingstone EJ, Cartwright JA, Campana L, Lewis PJS, Dwyer BJ, Aird R, Man TY, Vermeren M, Rossi AG, Boulter L, Forbes SJ. Semaphorin 7a is protective through immune modulation during acetaminophen-induced liver injury. J Inflamm (Lond) 2025; 22:13. [PMID: 40114253 PMCID: PMC11927371 DOI: 10.1186/s12950-025-00429-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/13/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND AND AIM Acetaminophen (APAP) induced acute liver injury (ALI), the leading cause acute liver failure in the western world, has limited treatment options. APAP toxicity results in massive hepatic necrosis and secondary infiltrating monocytes and neutrophils, which contribute to pathogenesis. Semaphorin 7a (Sema7a), a chemoattractant and modulator of monocytes and neutrophils, is a potential therapeutic target in other conditions, but its role in APAP-ALI is unexplored. METHODS Wild-type (WT) and Sema7a knockout (KO) mice were examined during APAP-ALI. Serum liver function tests, histological analysis and cellular localisation of Sema7a and its receptors, Plexin C1 and Integrin β1, were examined. Serum cytokines were quantified, tissue macrophages and neutrophils were localised, and in vivo phenotype, including phagocytosis, was assessed by immunohistochemistry and flow cytometry. RESULTS Sema7a was expressed by HNF4α + peri-necrotic hepatocytes circumferentially during APAP-ALI injury phases, and serum concentrations were increased, and correlated with hepatic injury. Sema7a KO mice had increased circulating inflammatory cytokines and significantly less hepatic F4/80 + macrophages, a cell type required for hepatic repair. Sema7a KO mice had higher necrotic area neutrophils, and increased neutrophil chemoattractant CXCL1. Without Sema7a expression, mice displayed increased necrosis and liver injury markers compared to Sema7a WT mice. Without peri-necrotic hepatocyte Sema7a expression, we also identified increased cell death and hepatic cellular stress outside of necrosis. CONCLUSION We have identified a novel protective role of Sema7a during injury phases of APAP-ALI. Without peri-necrotic hepatocyte Sema7a expression and secretion, there is increased inflammation, time specific worsened hepatic necrosis and increased hepatic cell stress and death outside of the necrotic zone.
Collapse
Affiliation(s)
- Eilidh J Livingstone
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Jennifer A Cartwright
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK.
- The Royal (Dick) School of Veterinary Studiesand Theaq , Roslin Institute, University of Edinburgh, Edinburgh, UK.
| | - Lara Campana
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Philip J Starkey Lewis
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Benjamin J Dwyer
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Rhona Aird
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Tak Yung Man
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Matthieu Vermeren
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Adriano Giorgio Rossi
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Luke Boulter
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Stuart John Forbes
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
5
|
Yang T, Zhang Y, Duan C, Liu H, Wang D, Liang Q, Chen X, Ma J, Cheng K, Chen Y, Zhuang R, Yin J. CD300E + macrophages facilitate liver regeneration after splenectomy in decompensated cirrhotic patients. Exp Mol Med 2025; 57:72-85. [PMID: 39741181 PMCID: PMC11799435 DOI: 10.1038/s12276-024-01371-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/15/2024] [Accepted: 09/30/2024] [Indexed: 01/02/2025] Open
Abstract
Liver cirrhosis is prognostically associated with poor life expectancy owing to subsequent liver failure. Thus, understanding liver regeneration processes during cirrhotic injury is highly important. This study explored the role of macrophage heterogeneity in liver regeneration following splenectomy. We collected detailed clinical information from 54 patients with decompensated cirrhosis before and after splenectomy. Obvious liver regeneration was observed after splenectomy in cirrhotic patients. Single-cell RNA sequencing (scRNA-seq) was performed on three paired liver tissues from patients before and after surgery to explore the immune microenvironment map and the characteristics of liver regeneration-associated macrophages (RAMs). scRNA-seq analysis revealed that the composition of hepatic immune cells changed after splenectomy; among these changes, the proportion of CD300E+ RAMs significantly increased after surgery, and high expression levels of functional genes associated with cell proliferation promoted liver regeneration. Moreover, a mouse model of carbon tetrachloride-induced cirrhosis and a coculture system consisting of primary bone marrow-derived macrophages and hepatocytes were established for validation. We observed a similar phenomenon of liver regeneration in cirrhotic mice and further confirmed that CD300E+ monocyte-derived macrophages facilitated hepatocyte NAD+ synthesis via the secretion of NAMPT, which subsequently promoted hepatocyte proliferation. This study characterized the hepatic immune microenvironment in patients with cirrhosis following splenectomy. Our findings demonstrated that CD300E+ macrophages play a crucial role in remodeling the hepatic immune microenvironment after splenectomy, thereby promoting liver regeneration in patients with decompensated cirrhosis. CD300E+ macrophages are anticipated to emerge as a novel therapeutic strategy for the treatment of liver cirrhosis.
Collapse
Affiliation(s)
- Tao Yang
- Department of General Surgery, Tangdu Hospital of the Air Force Medical University, 569 Xin Si Road, Xi'an, 710038, Shaanxi, China
| | - Yuan Zhang
- Department of Immunology, Air Force Medical University, 169 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Chujun Duan
- Department of Immunology, Air Force Medical University, 169 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Hui Liu
- Department of General Surgery, Tangdu Hospital of the Air Force Medical University, 569 Xin Si Road, Xi'an, 710038, Shaanxi, China
| | - Dong Wang
- Department of General Surgery, Tangdu Hospital of the Air Force Medical University, 569 Xin Si Road, Xi'an, 710038, Shaanxi, China
| | - Qingshan Liang
- Department of General Surgery, Tangdu Hospital of the Air Force Medical University, 569 Xin Si Road, Xi'an, 710038, Shaanxi, China
| | - Xiao Chen
- Department of General Surgery, Tangdu Hospital of the Air Force Medical University, 569 Xin Si Road, Xi'an, 710038, Shaanxi, China
| | - Jingchang Ma
- Department of Immunology, Air Force Medical University, 169 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Kun Cheng
- Department of Immunology, Air Force Medical University, 169 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Yong Chen
- Department of Hepatobiliary Surgery, Xijing Hospital of the Air Force Medical University, 15 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Ran Zhuang
- Department of Immunology, Air Force Medical University, 169 West Changle Road, Xi'an, 710032, Shaanxi, China.
| | - Jikai Yin
- Department of General Surgery, Tangdu Hospital of the Air Force Medical University, 569 Xin Si Road, Xi'an, 710038, Shaanxi, China.
| |
Collapse
|
6
|
Zhang X, Zhang F, Li M, Sun Q, Li Y, Fu Y, Zhang Y. Epicatechin attenuates emamectin benzoate-induced liver injury in grass carp by activating Nrf2/GPX4 signaling pathway. FISH & SHELLFISH IMMUNOLOGY 2025; 157:110118. [PMID: 39809417 DOI: 10.1016/j.fsi.2025.110118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 01/04/2025] [Accepted: 01/07/2025] [Indexed: 01/16/2025]
Abstract
Emamectin benzoate (EMB) is an antibiotic insecticide pesticide modified from avermectin. In the current study, we performed an in-depth investigation of the protective effects of epicatechin on EMB-induced liver injury in common carps. The carps were cultured in an aquatic environment containing 2.4 μg/L of EMB for 30 days to establish the model. The results provide direct evidence that epicatechin could attenuate EMB-induced liver injury, as confirmed by the inhibition of epicatechin on EMB-induced liver pathological injury, serum glucose (GLU), cortisol (COR), aspartate aminotransferase (AST), alanine aminotransferase (ALT), adenosine deaminase (ADA), and alkaline phosphatase (ALP) levels. Epicatechin also inhibited EMB-induced inflammation, as confirmed by the inhibition of epicatechin on tumor necrosis factor (TNF-α) and Interleukin-1β (IL-1β) production, and nuclear factor kappa-B (NF-κB) activation induced by EMB. Moreover, epicatechin could attenuate EMB-induced ferroptosis, as confirmed by the inhibition of epicatechin on malondialdehyde (MDA) and Fe2+ production, and up-regulation of Adenosine Triphosphate (ATP), Glutathione (GSH) production, and Glutathione Peroxidase 4 (Gpx4) and xCT expression. In addition, epicatechin increased the expression of nuclear factor erythroid-2 related factor 2 (Nrf2) and hemeoxygenase-1 (HO-1). We therefore conclude that epicatechin protected EMB-induced liver injury by preventing ferroptosis through activating Nrf2.
Collapse
Affiliation(s)
- Xinhui Zhang
- College of Animal Science and Technology, Jilin Agriculture Science and Technology University, Jilin City, China
| | - Fengyan Zhang
- College of Animal Science and Technology, Jilin Agriculture Science and Technology University, Jilin City, China
| | - Musen Li
- College of Animal Science and Technology, Jilin Agriculture Science and Technology University, Jilin City, China
| | - Qingsong Sun
- College of Animal Science and Technology, Jilin Agriculture Science and Technology University, Jilin City, China
| | - Yuehong Li
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, China; College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Yunhe Fu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, 130062, Jilin Province, China
| | - Yue Zhang
- College of Animal Science and Technology, Jilin Agriculture Science and Technology University, Jilin City, China.
| |
Collapse
|
7
|
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
|
8
|
Xie Z, Jiang J, Yang F, Han J, Ma Z, Wen T, Bai X. The C3/C3aR pathway exacerbates acetaminophen-induced mouse liver injury via upregulating podoplanin on the macrophage. FASEB J 2025; 39:e70272. [PMID: 39777689 PMCID: PMC11706223 DOI: 10.1096/fj.202402278rr] [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: 09/23/2024] [Revised: 11/26/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025]
Abstract
Acute liver failure (ALF) is a life-threatening condition that occurs when the liver sustains severe damage and rapidly loses its function. The primary cause of ALF is the overdose of acetaminophen (APAP), and its treatment is relatively limited. The involvement of the complement system in the development of ALF has been implicated. However, the related mechanisms remain poorly understood. Complement 3 (C3) knockout mice, complement 3a receptor (C3aR) knockout mice, platelet C-type lectin-like receptor 2 (Clec-2)-deficient mice, and myeloid cell podoplanin (Pdpn)-deficient mice were generated. Liver tissues were collected for histological analysis, RNA sequencing, confocal immunofluorescence, and immunoblot analyses. Our data demonstrated that APAP activated the C3/C3aR pathway, leading to intrahepatic hemorrhage, ultimately resulting in hepatocyte necrosis. Deletion of C3 or C3aR mitigated APAP-induced liver injury (AILI). C3/C3aR signaling upregulated the expression and phosphorylation of transcription factors STAT3 and c-Fos in hepatic Kupffer cells, which in turn increased PDPN expression, promoting platelet recruitment to the Kupffer cells via the interaction of PDPN and the CLEC-2 on platelets. Since the activation of platelets mediated by C3/C3aR occurs irrespective of the major hemostatic pathways, blocking the C3/C3aR pathway in ALF could be a coagulopathy-sparing and novel therapeutic approach. In summary, this study unveiled the critical roles of the C3/C3aR pathway in developing AILI, providing evidence that the C3/C3aR pathway could be an effective therapeutic target for AILI.
Collapse
Affiliation(s)
- Zhanli Xie
- Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, NHC Key Laboratory of Thrombosis and HemostasisThe First Affiliated Hospital of Soochow UniversitySuzhouChina
- Institute of Clinical Medicine Research, Suzhou Hospital, Affiliated Hospital of Medical SchoolNanjing UniversitySuzhouChina
| | - Jiang Jiang
- Department of Nuclear MedicineThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Fei Yang
- Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, NHC Key Laboratory of Thrombosis and HemostasisThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Jingjing Han
- Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, NHC Key Laboratory of Thrombosis and HemostasisThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Zhenni Ma
- Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, NHC Key Laboratory of Thrombosis and HemostasisThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Tao Wen
- Medical Research CenterBeijing Chao‐Yang Hospital, Capital Medical UniversityBeijingChina
| | - Xia Bai
- Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, NHC Key Laboratory of Thrombosis and HemostasisThe First Affiliated Hospital of Soochow UniversitySuzhouChina
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of HematologySoochow UniversitySuzhouChina
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouChina
| |
Collapse
|
9
|
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
|
10
|
Bukhari I, Li M, Li G, Xu J, Zheng P, Chu X. Pinpointing the integration of artificial intelligence in liver cancer immune microenvironment. Front Immunol 2024; 15:1520398. [PMID: 39759506 PMCID: PMC11695355 DOI: 10.3389/fimmu.2024.1520398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 12/02/2024] [Indexed: 01/07/2025] Open
Abstract
Liver cancer remains one of the most formidable challenges in modern medicine, characterized by its high incidence and mortality rate. Emerging evidence underscores the critical roles of the immune microenvironment in tumor initiation, development, prognosis, and therapeutic responsiveness. However, the composition of the immune microenvironment of liver cancer (LC-IME) and its association with clinicopathological significance remain unelucidated. In this review, we present the recent developments related to the use of artificial intelligence (AI) for studying the immune microenvironment of liver cancer, focusing on the deciphering of complex high-throughput data. Additionally, we discussed the current challenges of data harmonization and algorithm interpretability for studying LC-IME.
Collapse
Affiliation(s)
- Ihtisham Bukhari
- Department of Oncology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Marshall B. J. Medical Research Center, Zhengzhou University, Zhengzhou, Henan, China
| | - Mengxue Li
- Marshall B. J. Medical Research Center, Zhengzhou University, Zhengzhou, Henan, China
| | - Guangyuan Li
- Department of Oncology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jixuan Xu
- Department of Gastrointestinal & Thyroid Surgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Pengyuan Zheng
- Marshall B. J. Medical Research Center, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiufeng Chu
- Department of Oncology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Marshall B. J. Medical Research Center, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
11
|
He J, Fu L, Shen Y, Teng Y, Huang Y, Ding X, Xu D, Cui H, Zhu M, Xie J, Su Y, Li T, Huang W, Mou X, Bian Q, Fan Y. Polygonum multiflorum Extracellular Vesicle-Like Nanovesicle for Skin Photoaging Therapy. Biomater Res 2024; 28:0098. [PMID: 39703536 PMCID: PMC11658808 DOI: 10.34133/bmr.0098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/13/2024] [Accepted: 10/08/2024] [Indexed: 12/21/2024] Open
Abstract
Ultraviolet (UV) irradiation leads to the degradation of the extracellular matrix and collagen, thereby accelerating skin aging and imposing substantial psychological burden on patients. Current anti-aging strategies are limited and often associated with high costs or strong side effects. Plant-derived extracellular vesicle-like nanovesicles, with advantages such as natural availability and cost-effectiveness, show potential in anti-aging interventions. This study extracted extracellular vesicle-like nanovesicle from Polygonum multiflorum (PMELNVs) and systematically investigated their composition and metabolic pathways, further examining their efficacy and underlying mechanisms in combating photoaging. Results revealed the excellent antioxidative properties of PMELNVs, alleviating UV-induced oxidative stress, inhibiting matrix metalloproteinase production, reducing extracellular matrix degradation, promoting collagen synthesis, and ultimately exerting anti-photoaging effects. Additionally, safety assessments demonstrated favorable biocompatibility of PMELNVs. This study provides novel evidence supporting PMELNVs' ability to resist photoaging by reducing oxidative stress and enhancing collagen expression, thereby offering potential as a new natural therapeutic agent against skin photoaging and promising a safer and more effective local anti-aging strategy.
Collapse
Affiliation(s)
- Junjia He
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital,
Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
- Clinical Research Institute, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital,
Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Luoqin Fu
- Clinical Research Institute, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital,
Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Yeyu Shen
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital,
Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
- Clinical Research Institute, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital,
Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Yan Teng
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital,
Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Youming Huang
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital,
Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Xiaoxia Ding
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital,
Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Danfeng Xu
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital,
Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Hong Cui
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital,
Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Mingang Zhu
- Department of Dermatology, the First People’s Hospital of Jiashan, Jiaxing, Zhejiang 314100, China
| | - Jiahao Xie
- The Second Clinical Medical College,
Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310014, China
| | - Yue Su
- The Second Clinical Medical College,
Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310014, China
| | - Ting Li
- College of Bioengineering,
Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Weitao Huang
- Clinical Research Institute, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital,
Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Xiaozhou Mou
- Clinical Research Institute, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital,
Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Qiong Bian
- Clinical Research Institute, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital,
Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Yibin Fan
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital,
Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| |
Collapse
|
12
|
Vandendriessche S, Mattos MS, Bialek EL, Schuermans S, Proost P, Marques PE. Complement activation drives the phagocytosis of necrotic cell debris and resolution of liver injury. Front Immunol 2024; 15:1512470. [PMID: 39759517 PMCID: PMC11696981 DOI: 10.3389/fimmu.2024.1512470] [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: 10/16/2024] [Accepted: 11/29/2024] [Indexed: 01/07/2025] Open
Abstract
Cells die by necrosis due to excessive chemical or thermal stress, leading to plasma membrane rupture, release of intracellular components and severe inflammation. The clearance of necrotic cell debris is crucial for tissue recovery and injury resolution, however, the underlying mechanisms are still poorly understood, especially in vivo. This study examined the role of complement proteins in promoting clearance of necrotic cell debris by leukocytes and their influence on liver regeneration. We found that independently of the type of necrotic liver injury, either acetaminophen (APAP) overdose or thermal injury, complement proteins C1q and (i)C3b were deposited specifically on necrotic lesions via the activation of the classical pathway. Importantly, C3 deficiency led to a significant accumulation of necrotic debris and impairment of liver recovery in mice, which was attributed to decreased phagocytosis of debris by recruited neutrophils in vivo. Monocytes and macrophages also took part in debris clearance, although the necessity of C3 and CD11b was dependent on the specific type of necrotic liver injury. Using human neutrophils, we showed that absence of C3 or C1q caused a reduction in the volume of necrotic debris that is phagocytosed, indicating that complement promotes effective debris uptake in mice and humans. Moreover, internalization of opsonized debris induced the expression of pro-resolving genes in a C3-dependent manner, supporting the notion that debris clearance favors the resolution of inflammation. In summary, complement activation at injury sites is a pivotal event for necrotic debris clearance by phagocytes and determinant for efficient recovery from tissue injury.
Collapse
Affiliation(s)
| | | | | | | | | | - Pedro Elias Marques
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
13
|
Rao J, Ye D, Ren A, He W, Zhang X, Chen P, Jian Q, Fu Z, Deng R, Hu Y, Gao Y, Ma Y. Macrophage Evolution during Progression of Hepatitis Virus B-Related Acute-on-Chronic Liver Failure. J Innate Immun 2024; 17:29-43. [PMID: 39637841 PMCID: PMC11753795 DOI: 10.1159/000542946] [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: 05/23/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024] Open
Abstract
INTRODUCTION Hepatitis B virus (HBV)-related liver diseases, including hepatitis, cirrhosis, and liver failure, seriously threaten human lives and health worldwide. Innate and adaptive immune cells are all thought to participate in HBV-related diseases. However, there is a lack of information on the comprehensive landscape of the immune microenvironment. METHODS In this study, single-cell ribonucleic acid sequencing was performed on liver samples obtained from patients diagnosed with hepatitis, cirrhosis, and acute-on-chronic liver failure, which were caused by HBV. Trajectory analysis was performed to analyze the evolution of cell subsets, and branch expression analysis modeling was applied to visualize the changes in gene expression during evolution. RESULTS Finally, there was a significant increase in adaptive immune cells in the hepatitis and cirrhosis groups, whereas more innate immune cells were observed in the liver failure group. Furthermore, we found that monocytes underwent remarkable transcriptomic changes into FABP5+ macrophages, promoting the degranulation and chemotaxis of neutrophils through RESISTIN signaling, and LGMN+ macrophages, with the sequential activation of antigen presentation and defense to pathogens through SPP1 signaling. CONCLUSION Macrophages were revealed as central to the progression of acute-on-chronic liver failure as they regulated the activation or inhibition of other immune cells, which could help in developing an effective novel therapy.
Collapse
Affiliation(s)
- Jiawei Rao
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
| | - Dongmei Ye
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
| | - Ao Ren
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Wenjin He
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
| | - Xuzhi Zhang
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
| | - Pengrui Chen
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
| | - Qian Jian
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
| | - Zongli Fu
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
| | - Ronghai Deng
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
| | - Yixin Hu
- State Key Laboratory of Oncology in South China, Department of Ultrasound Sun Yat-Sen University Cancer Center, and Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Yifang Gao
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
| | - Yi Ma
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
| |
Collapse
|
14
|
Ni Q, Lin J, Huang W, Yang L, Li R, Tu T, He G, Gao Y, Sun X, Kong X, Zhu X. Kehuang capsule inhibits MAPK and AKT signaling pathways to mitigate CCl 4-induced acute liver injury. LIVER RESEARCH 2024; 8:269-281. [PMID: 39958915 PMCID: PMC11771260 DOI: 10.1016/j.livres.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/27/2024] [Accepted: 11/28/2024] [Indexed: 02/18/2025]
Abstract
Background and aims Kehuang (KH) capsule is an herbal medical product approved for the treatment of liver diseases, including liver injury, in China. However, the mechanism is still unclear. This study aimed to elucidate the protective effects of KH capsule against carbon tetrachloride (CCl4)-induced acute liver injury (ALI) in a murine model. Methods Mice were randomly divided into control, model (CCl4), CCl4+KH_Low and CCl4+KH_High group. Liver enzyme levels and histological changes were assessed to evaluate liver injury. Oxidative stress markers and inflammatory cell infiltration in liver tissues were measured. Additionally, network pharmacology was employed to explore the potential mechanisms of KH capsule. Results KH capsule significantly reduced serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels, as well as the necrotic area in liver tissue. KH capsule also decreased the infiltration of macrophages and neutrophils, thereby inhibiting the expression of interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), and interleukin-1 beta (IL-1β). Furthermore, KH capsule decreased liver malondialdehyde (MDA) levels and increased superoxide dismutase (SOD) activity. The number of terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick-end labeling (TUNEL)-positive cells in liver tissue was also reduced. The expression of nuclear factor erythroid 2 related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) proteins was significantly elevated, while the protein expression of cytochrome P450 2E1 (CYP2E1) was significantly reduced. Mass spectrometry identified genistein, galangin, wogonin, skullcapflavone II, and hispidulin as potential active ingredients of KH capsule. Network pharmacology analysis revealed enrichment in the mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3K)-protein kinase B (AKT) signaling pathways. Western blot analysis confirmed that KH capsule suppressed AKT, extracellular signal-regulated kinase (ERK), and p38 signaling. Conclusions These findings suggest that KH capsule could exert protective effects against CCl4-induced ALI, with the inhibition of MAPK and PI3K-AKT signaling pathways playing a crucial role in its mechanism of action.
Collapse
Affiliation(s)
- Qinyu Ni
- Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Liver Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiacheng Lin
- Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weifan Huang
- Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liu Yang
- Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ran Li
- Kexing Biopharm Co., Ltd, Jinan, Shandong, China
| | - Tianzhi Tu
- Kexing Biopharm Co., Ltd, Jinan, Shandong, China
| | - Guangfu He
- Kexing Biopharm Co., Ltd, Jinan, Shandong, China
| | - Yueqiu Gao
- Department of Liver Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xuehua Sun
- Department of Liver Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoni Kong
- Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaojun Zhu
- Department of Liver Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
15
|
Zheng J, Xiao J, Fan Y, Zheng H, Liu H, Xiang J, Hai L, Wang Y, Zhang X. CD24 regulates liver immune response and ameliorates acute hepatic injury through controlling hepatic macrophages. Eur J Immunol 2024; 54:e2451178. [PMID: 39444061 DOI: 10.1002/eji.202451178] [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: 04/07/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/25/2024]
Abstract
Liver injury releases danger-associated molecular patterns, which trigger the immune response. CD24 negatively regulates the immune response by binding with danger-associated molecular patterns, but the specific role of CD24 in modulating macrophage-related inflammation during liver injury remains largely unexplored. Here, we aimed to investigate the mechanisms of macrophage CD24 in the development of liver injury. Our results show that CD24 expression is upregulated primarily in hepatic macrophages (HMs) during acute liver injury. CD24-deficient mice exhibited more severe liver injury and showed a significantly higher frequency and number of HMs, particularly Ly6Chi monocyte-derived macrophages. Mechanistically, the CD24-Siglec-G interaction plays a vital role in mitigating acute liver injury. CD24-mediated inhibitory signaling in HMs primarily limits downstream NF-κB and p38 MAPK activation through the recruitment of SHP1. Our work unveils the critical role of macrophage CD24 in negatively regulating innate immune responses and protecting against acute liver injury, thus providing potential therapeutic targets for liver-associated diseases.
Collapse
Affiliation(s)
- Jian Zheng
- Key Laboratory of Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, P. R. China
| | - Jun Xiao
- Key Laboratory of Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, P. R. China
| | - Yatong Fan
- Department of Blood Transfusion, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, P. R. China
| | - Honggang Zheng
- Department of Pathology, Tianjin Jinyu Medical Laboratory Co LTD, Tianjin, P. R. China
| | - Hongyu Liu
- Key Laboratory of Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, P. R. China
| | - Jie Xiang
- Key Laboratory of Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, P. R. China
| | - Lei Hai
- Key Laboratory of Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, P. R. China
| | - Yan Wang
- Key Laboratory of Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, P. R. China
| | - Xuejun Zhang
- Key Laboratory of Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, P. R. China
| |
Collapse
|
16
|
Li Q, Xu Q, Shi J, Dong W, Jin J, Zhang C. FAK inhibition delays liver repair after acetaminophen-induced acute liver injury by suppressing hepatocyte proliferation and macrophage recruitment. Hepatol Commun 2024; 8:e0531. [PMID: 39761008 PMCID: PMC11495758 DOI: 10.1097/hc9.0000000000000531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 08/13/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND Overdose of acetaminophen (APAP), a commonly used antipyretic analgesic, can lead to severe liver injury and failure. Current treatments are only effective in the early stages of APAP-induced acute liver injury (ALI). Therefore, a detailed examination of the mechanisms involved in liver repair following APAP-induced ALI could provide valuable insights for clinical interventions. METHODS 4D-label-free proteomics analysis was used to identify dysregulated proteins in the liver of APAP-treated mice. RNA-Seq, hematoxylin-eosin staining, immunohistochemical staining, immunofluorescence staining, quantitative PCR, western blotting, transwell were used to explore the underlying mechanisms. RESULTS Utilizing high throughput 4D-label-free proteomics analysis, we observed a notable increase in proteins related to the "focal adhesion" pathway in the livers of APAP-treated mice. Inhibiting focal adhesion kinase (FAK) activation with a specific inhibitor, 1,2,4,5-Benzenetetraamine tetrahydrochloride (also called Y15), resulted in reduced macrophage numbers, delayed necrotic cell clearance, and inhibited liver cell proliferation in the necrotic regions of APAP-treated mice. RNA-Seq analysis demonstrated that Y15 downregulated genes associated with "cell cycle" and "phagosome" pathways in the livers of APAP-treated mice. Furthermore, blocking extracellular matrix (ECM)-integrin activation with a competitive peptide inhibitor, Gly-Arg-Gly-Asp-Ser (GRGDS), suppressed FAK activation and liver cell proliferation without affecting macrophage recruitment to necrotic areas. Mechanistically, ECM-induced FAK activation upregulated growth-promoting cell cycle genes, leading to hepatocyte proliferation, while CCL2 enhanced FAK activation and subsequent macrophage recruitment via F-actin rearrangement. CONCLUSIONS Overall, these findings underscore the pivotal role of FAK activation in liver repair post-APAP overdose by promoting liver cell proliferation and macrophage recruitment.
Collapse
Affiliation(s)
- Qing Li
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
- China-USA Lipids in Health and Disease Research Center, Guilin Medical University, Guilin, Guangxi, China
| | - Qi Xu
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Jialin Shi
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Wei Dong
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Junfei Jin
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
- China-USA Lipids in Health and Disease Research Center, Guilin Medical University, Guilin, Guangxi, China
| | - Chong Zhang
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
- China-USA Lipids in Health and Disease Research Center, Guilin Medical University, Guilin, Guangxi, China
| |
Collapse
|
17
|
Tanabe M, Hosono K, Yamashita A, Ito Y, Majima M, Narumiya S, Kusano C, Amano H. Deletion of TP signaling in macrophages delays liver repair following APAP-induced liver injury by reducing accumulation of reparative macrophage and production of HGF. Inflamm Regen 2024; 44:43. [PMID: 39363292 PMCID: PMC11451145 DOI: 10.1186/s41232-024-00356-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 09/26/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND Acetaminophen (APAP)-induced liver injury is the most common cause of acute liver failure. Macrophages are key players in liver restoration following APAP-induced liver injury. Thromboxane A2 (TXA2) and its receptor, thromboxane prostanoid (TP) receptor, have been shown to be involved in tissue repair. However, whether TP signaling plays a role in liver repair after APAP hepatotoxicity by affecting macrophage function remains unclear. METHODS Male TP knockout (TP-/-) and C57BL/6 wild-type (WT) mice were treated with APAP (300 mg/kg). In addition, macrophage-specific TP-knockout (TP△mac) and control WT mice were treated with APAP. We explored changes in liver inflammation, liver repair, and macrophage accumulation in mice treated with APAP. RESULTS Compared with WT mice, TP-/- mice showed aggravated liver injury as indicated by increased levels of alanine transaminase (ALT) and necrotic area as well as delayed liver repair as indicated by decreased expression of proliferating cell nuclear antigen (PCNA). Macrophage deletion exacerbated APAP-induced liver injury and impaired liver repair. Transplantation of TP-deficient bone marrow (BM) cells to WT or TP-/- mice aggravated APAP hepatotoxicity with suppressed accumulation of macrophages, while transplantation of WT-BM cells to WT or TP-/- mice attenuated APAP-induced liver injury with accumulation of macrophages in the injured regions. Macrophage-specific TP-/- mice exacerbated liver injury and delayed liver repair, which was associated with increased pro-inflammatory macrophages and decreased reparative macrophages and hepatocyte growth factor (HGF) expression. In vitro, TP signaling facilitated macrophage polarization to a reparative phenotype. Transfer of cultured BM-derived macrophages from control mice to macrophage-specific TP-/- mice attenuated APAP-induced liver injury and promoted liver repair. HGF treatment mitigated APAP-induced inflammation and promoted liver repair after APAP-induced liver injury. CONCLUSIONS Deletion of TP signaling in macrophages delays liver repair following APAP-induced liver injury, which is associated with reduced accumulation of reparative macrophages and the hepatotrophic factor HGF. Specific activation of TP signaling in macrophages may be a potential therapeutic target for liver repair and regeneration after APAP hepatotoxicity.
Collapse
Affiliation(s)
- Mina Tanabe
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Japan
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Japan
- Department of Gastroenterology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Kanako Hosono
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Japan
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Japan
| | - Atsushi Yamashita
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Japan
| | - Yoshiya Ito
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Japan
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Japan
| | - Masataka Majima
- Department of Medical Therapeutics, Kanagawa Institute of Technology, Atsugi, Japan
| | - Shuh Narumiya
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Chika Kusano
- Department of Gastroenterology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Hideki Amano
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Japan.
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Japan.
| |
Collapse
|
18
|
Hinz K, Niu M, Ni HM, Ding WX. Targeting Autophagy for Acetaminophen-Induced Liver Injury: An Update. LIVERS 2024; 4:377-387. [PMID: 39301093 PMCID: PMC11412313 DOI: 10.3390/livers4030027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/22/2024] Open
Abstract
Acetaminophen (APAP) overdose can induce hepatocyte necrosis and acute liver failure in experimental rodents and humans. APAP is mainly metabolized via hepatic cytochrome P450 enzymes to generate the highly reactive metabolite N-acetyl-p-benzoquinone imine (NAPQI), which forms acetaminophen protein adducts (APAP-adducts) and damages mitochondria, triggering necrosis. APAP-adducts and damaged mitochondria can be selectively removed by autophagy. Increasing evidence implies that the activation of autophagy may be beneficial for APAP-induced liver injury (AILI). In this minireview, we briefly summarize recent progress on autophagy, in particular, the pharmacological targeting of SQSTM1/p62 and TFEB in AILI.
Collapse
Affiliation(s)
- Kaitlyn Hinz
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Mengwei Niu
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Hong-Min Ni
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
19
|
Luyendyk JP, Morozova E, Copple BL. Good Cells Go Bad: Immune Dysregulation in the Transition from Acute Liver Injury to Liver Failure After Acetaminophen Overdose. Drug Metab Dispos 2024; 52:722-728. [PMID: 38050055 PMCID: PMC11257689 DOI: 10.1124/dmd.123.001280] [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: 09/11/2023] [Revised: 11/12/2023] [Accepted: 11/27/2023] [Indexed: 12/06/2023] Open
Abstract
The role of inflammatory cells and other components of the immune system in acetaminophen (APAP)-induced liver injury and repair has been extensively investigated. Although this has resulted in a wealth of information regarding the function and regulation of immune cells in the liver after injury, apparent contradictions have fueled controversy around the central question of whether the immune system is beneficial or detrimental after APAP overdose. Ultimately, this may not be a simple assignment of "good" or "bad." Clinical studies have clearly demonstrated an association between immune dysregulation and a poor outcome in patients with severe liver damage/liver failure induced by APAP overdose. To date, studies in mice have not uniformly replicated this connection. The apparent disconnect between clinical and experimental studies has perhaps stymied progress and further complicated investigation of the immune system in APAP-induced liver injury. Mouse models are often dismissed as not recapitulating the clinical scenario. Moreover, clinical investigation is most often focused on the most severe APAP overdose patients, those with liver failure. Notably, recent studies have made it apparent that the functional role of the immune system in the pathogenesis of APAP-induced liver injury is highly context dependent and greatly influenced by the experimental conditions. In this review, we highlight some of these recent findings and suggest strategies seeking to resolve and build on existing disconnects in the literature. SIGNIFICANCE STATEMENT: Acetaminophen overdose is the most frequent cause of acute liver failure in the United States. Studies indicate that dysregulated innate immunity contributes to the transition from acute liver injury to acute liver failure. In this review, we discuss the evidence for this and the potential underlying causes.
Collapse
Affiliation(s)
- James P Luyendyk
- Departments of Pathobiology and Diagnostic Investigation (J.P.L., E.M.) and Pharmacology and Toxicology (B.L.C.), Michigan State University, East Lansing, Michigan
| | - Elena Morozova
- Departments of Pathobiology and Diagnostic Investigation (J.P.L., E.M.) and Pharmacology and Toxicology (B.L.C.), Michigan State University, East Lansing, Michigan
| | - Bryan L Copple
- Departments of Pathobiology and Diagnostic Investigation (J.P.L., E.M.) and Pharmacology and Toxicology (B.L.C.), Michigan State University, East Lansing, Michigan
| |
Collapse
|
20
|
McGill MR. The Role of Mechanistic Biomarkers in Understanding Acetaminophen Hepatotoxicity in Humans. Drug Metab Dispos 2024; 52:729-739. [PMID: 37918967 PMCID: PMC11257692 DOI: 10.1124/dmd.123.001281] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/18/2023] [Accepted: 10/30/2023] [Indexed: 11/04/2023] Open
Abstract
Our understanding of the fundamental molecular mechanisms of acetaminophen (APAP) hepatotoxicity began in 1973 to 1974, when investigators at the US National Institutes of Health published seminal studies demonstrating conversion of APAP to a reactive metabolite that depletes glutathione and binds to proteins in the liver in mice after overdose. Since then, additional groundbreaking experiments have demonstrated critical roles for mitochondrial damage, oxidative stress, nuclear DNA fragmentation, and necrotic cell death as well. Over the years, some investigators have also attempted to translate these mechanisms to humans using human specimens from APAP overdose patients. This review presents those studies and summarizes what we have learned about APAP hepatotoxicity in humans so far. Overall, the mechanisms of APAP hepatotoxicity in humans strongly resemble those discovered in experimental mouse and cultured hepatocyte models, and emerging biomarkers also suggest similarities in liver repair. The data not only validate the first mechanistic studies of APAP-induced liver injury performed 50 years ago but also demonstrate the human relevance of numerous studies conducted since then. SIGNIFICANCE STATEMENT: Human studies using novel translational, mechanistic biomarkers have confirmed that the fundamental mechanisms of acetaminophen (APAP) hepatotoxicity discovered in rodent models since 1973 are the same in humans. Importantly, these findings have guided the development and understanding of treatments such as N-acetyl-l-cysteine and 4-methylpyrazole over the years. Additional research may improve not only our understanding of APAP overdose pathophysiology in humans but also our ability to predict and treat serious liver injury in patients.
Collapse
Affiliation(s)
- Mitchell R McGill
- Department of Environmental Health Sciences, Fay W. Boozman College of Public Health; Department of Pharmacology and Toxicology, College of Medicine; and Department of Pathology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| |
Collapse
|
21
|
Sugino KY, Janssen RC, McMahan RH, Zimmerman C, Friedman JE, Jonscher KR. Vertical Transfer of Maternal Gut Microbes to Offspring of Western Diet-Fed Dams Drives Reduced Levels of Tryptophan Metabolites and Postnatal Innate Immune Response. Nutrients 2024; 16:1808. [PMID: 38931163 PMCID: PMC11206590 DOI: 10.3390/nu16121808] [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: 05/09/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Maternal obesity and/or Western diet (WD) is associated with an increased risk of metabolic dysfunction-associated steatotic liver disease (MASLD) in offspring, driven, in part, by the dysregulation of the early life microbiome. Here, using a mouse model of WD-induced maternal obesity, we demonstrate that exposure to a disordered microbiome from WD-fed dams suppressed circulating levels of endogenous ligands of the aryl hydrocarbon receptor (AHR; indole, indole-3-acetate) and TMAO (a product of AHR-mediated transcription), as well as hepatic expression of Il10 (an AHR target), in offspring at 3 weeks of age. This signature was recapitulated by fecal microbial transfer from WD-fed pregnant dams to chow-fed germ-free (GF) lactating dams following parturition and was associated with a reduced abundance of Lactobacillus in GF offspring. Further, the expression of Il10 was downregulated in liver myeloid cells and in LPS-stimulated bone marrow-derived macrophages (BMDM) in adult offspring, suggestive of a hypo-responsive, or tolerant, innate immune response. BMDMs from adult mice lacking AHR in macrophages exhibited a similar tolerogenic response, including diminished expression of Il10. Overall, our study shows that exposure to maternal WD alters microbial metabolites in the offspring that affect AHR signaling, potentially contributing to innate immune hypo-responsiveness and progression of MASLD, highlighting the impact of early life gut dysbiosis on offspring metabolism. Further investigations are warranted to elucidate the complex interplay between maternal diet, gut microbial function, and the development of neonatal innate immune tolerance and potential therapeutic interventions targeting these pathways.
Collapse
Affiliation(s)
- Kameron Y. Sugino
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.Y.S.); (R.C.J.); (J.E.F.)
| | - Rachel C. Janssen
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.Y.S.); (R.C.J.); (J.E.F.)
| | - Rachel H. McMahan
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Chelsea Zimmerman
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Jacob E. Friedman
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.Y.S.); (R.C.J.); (J.E.F.)
- Department of Biochemistry and Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Karen R. Jonscher
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.Y.S.); (R.C.J.); (J.E.F.)
- Department of Biochemistry and Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
22
|
Abstract
Acute liver injury (ALI), that is, the development of reduced liver function in patients without preexisting liver disease, can result from a wide range of causes, such as viral or bacterial infection, autoimmune disease, or adverse reaction to prescription and over-the-counter medications. ALI patients present with a complex coagulopathy, characterized by both hypercoagulable and hypocoagulable features. Similarly, ALI patients display a profound dysregulation of the fibrinolytic system with the vast majority of patients presenting with a hypofibrinolytic phenotype. Decades of research in experimental acute liver injury in mice suggest that fibrinolytic proteins, including plasmin(ogen), plasminogen activators, fibrinolysis inhibitors, and fibrin(ogen), can contribute to initial hepatotoxicity and/or stimulate liver repair. This review summarizes major experimental findings regarding the role of fibrinolytic factors in ALI from the last approximately 30 years and identifies unanswered questions, as well as highlighting areas for future research.
Collapse
Affiliation(s)
- Gina E Capece
- Department of Pharmacology, Rutgers University Robert Wood Johnson Medical School, Piscataway, New Jersey
| | - James P Luyendyk
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan
| | - Lauren G Poole
- Department of Pharmacology, Rutgers University Robert Wood Johnson Medical School, Piscataway, New Jersey
| |
Collapse
|
23
|
Adelusi OB, Akakpo JY, Eichenbaum G, Sadaff E, Ramachandran A, Jaeschke H. The thrombopoietin mimetic JNJ-26366821 reduces the late injury and accelerates the onset of liver recovery after acetaminophen-induced liver injury in mice. Arch Toxicol 2024; 98:1843-1858. [PMID: 38551724 PMCID: PMC11210275 DOI: 10.1007/s00204-024-03725-2] [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: 10/21/2023] [Accepted: 03/04/2024] [Indexed: 05/21/2024]
Abstract
Acetaminophen (APAP)-induced hepatotoxicity is comprised of an injury and recovery phase. While pharmacological interventions, such as N-acetylcysteine (NAC) and 4-methylpyrazole (4-MP), prevent injury there are no therapeutics that promote recovery. JNJ-26366821 (TPOm) is a novel thrombopoietin mimetic peptide with no sequence homology to endogenous thrombopoietin (TPO). Endogenous thrombopoietin is produced by hepatocytes and the TPO receptor is present on liver sinusoidal endothelial cells in addition to megakaryocytes and platelets, and we hypothesize that TPOm activity at the TPO receptor in the liver provides a beneficial effect following liver injury. Therefore, we evaluated the extent to which TPOm, NAC or 4-MP can provide a protective and regenerative effect in the liver when administered 2 h after an APAP overdose of 300 mg/kg in fasted male C57BL/6J mice. TPOm did not affect protein adducts, oxidant stress, DNA fragmentation and hepatic necrosis up to 12 h after APAP. In contrast, TPOm treatment was beneficial at 24 h, i.e., all injury parameters were reduced by 42-48%. Importantly, TPOm enhanced proliferation by 100% as indicated by PCNA-positive hepatocytes around the area of necrosis. When TPOm treatment was delayed by 6 h, there was no effect on the injury, but a proliferative effect was still evident. In contrast, 4MP and NAC treated at 2 h after APAP significantly attenuated all injury parameters at 24 h but failed to enhance hepatocyte proliferation. Thus, TPOm arrests the progression of liver injury by 24 h after APAP and accelerates the onset of the proliferative response which is essential for liver recovery.
Collapse
Affiliation(s)
- Olamide B Adelusi
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd, MS 1018, Kansas City, KS, 66160, USA
| | - Jephte Y Akakpo
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd, MS 1018, Kansas City, KS, 66160, USA
| | - Gary Eichenbaum
- Office of the Chief Medical Officer, Johnson & Johnson, Consumer Health, New Brunswick, NJ, 08901, USA
| | - Ejaz Sadaff
- Office of the Chief Medical Officer, Johnson & Johnson, Consumer Health, New Brunswick, NJ, 08901, USA
| | - Anup Ramachandran
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd, MS 1018, Kansas City, KS, 66160, USA
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd, MS 1018, Kansas City, KS, 66160, USA.
| |
Collapse
|
24
|
Ligeron C, Saenz J, Evrard B, Drouin M, Merieau E, Mary C, Biteau K, Wilhelm E, Batty C, Gauttier V, Baccelli I, Poirier N, Chiffoleau E. CLEC-1 Restrains Acute Inflammatory Response and Recruitment of Neutrophils following Tissue Injury. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1178-1187. [PMID: 38353642 DOI: 10.4049/jimmunol.2300479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 01/17/2024] [Indexed: 03/20/2024]
Abstract
The inflammatory response is a key mechanism for the elimination of injurious agents but must be tightly controlled to prevent additional tissue damage and progression to persistent inflammation. C-type lectin receptors expressed mostly by myeloid cells play a crucial role in the regulation of inflammation by recognizing molecular patterns released by injured tissues. We recently showed that the C-type lectin receptor CLEC-1 is able to recognize necrotic cells. However, its role in the acute inflammatory response following tissue damage had not yet been investigated. We show in this study, in a mouse model of liver injury induced by acetaminophen intoxication, that Clec1a deficiency enhances the acute immune response with increased expression of Il1b, Tnfa, and Cxcl2 and higher infiltration of activated neutrophils into the injured organ. Furthermore, we demonstrate that Clec1a deficiency exacerbates tissue damage via CXCL2-dependent neutrophil infiltration. In contrast, we observed that the lack of CLEC-1 limits CCL2 expression and the accumulation, beyond the peak of injury, of monocyte-derived macrophages. Mechanistically, we found that Clec1a-deficient dendritic cells increase the expression of Il1b, Tnfa, and Cxcl2 in response to necrotic cells, but decrease the expression of Ccl2. Interestingly, treatment with an anti-human CLEC-1 antagonist mAb recapitulates the exacerbation of acute immunopathology observed by genetic loss of Clec1a in a preclinical humanized mouse model. To conclude, our results demonstrate that CLEC-1 is a death receptor limiting the acute inflammatory response following injury and represents a therapeutic target to modulate immunity.
Collapse
Affiliation(s)
- Camille Ligeron
- OSE Immunotherapeutics, Nantes, France
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Javier Saenz
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Berangere Evrard
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Marion Drouin
- OSE Immunotherapeutics, Nantes, France
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Emmanuel Merieau
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | | | | | | | | | | | | | | | - Elise Chiffoleau
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| |
Collapse
|
25
|
Liu FC, Yu HP, Lee HC, Chen CY, Liao CC. The Modulation of Phospho-Extracellular Signal-Regulated Kinase and Phospho-Protein Kinase B Signaling Pathways plus Activity of Macrophage-Stimulating Protein Contribute to the Protective Effect of Stachydrine on Acetaminophen-Induced Liver Injury. Int J Mol Sci 2024; 25:1484. [PMID: 38338766 PMCID: PMC10855734 DOI: 10.3390/ijms25031484] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/16/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
Stachydrine, a prominent bioactive alkaloid derived from Leonurus heterophyllus, is a significant herb in traditional medicine. It has been noted for its anti-inflammatory and antioxidant characteristics. Consequently, we conducted a study of its hepatoprotective effect and the fundamental mechanisms involved in acetaminophen (APAP)-induced liver injury, utilizing a mouse model. Mice were intraperitoneally administered a hepatotoxic dose of APAP (300 mg/kg). Thirty minutes after APAP administration, mice were treated with different concentrations of stachydrine (0, 2.5, 5, and 10 mg/kg). Animals were sacrificed 16 h after APAP injection for serum and liver tissue assays. APAP overdose significantly elevated the serum alanine transferase levels, hepatic pro-inflammatory cytokines, malondialdehyde activity, phospho-extracellular signal-regulated kinase (ERK), phospho-protein kinase B (AKT), and macrophage-stimulating protein expression. Stachydrine treatment significantly decreased these parameters in mice with APAP-induced liver damage. Our results suggest that stachydrine may be a promising beneficial target in the prevention of APAP-induced liver damage through attenuation of the inflammatory response, inhibition of the ERK and AKT pathways, and expression of macrophage-stimulating proteins.
Collapse
Affiliation(s)
- Fu-Chao Liu
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (F.-C.L.); (H.-P.Y.); (H.-C.L.); (C.-Y.C.)
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Huang-Ping Yu
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (F.-C.L.); (H.-P.Y.); (H.-C.L.); (C.-Y.C.)
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Hung-Chen Lee
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (F.-C.L.); (H.-P.Y.); (H.-C.L.); (C.-Y.C.)
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chun-Yu Chen
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (F.-C.L.); (H.-P.Y.); (H.-C.L.); (C.-Y.C.)
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chia-Chih Liao
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (F.-C.L.); (H.-P.Y.); (H.-C.L.); (C.-Y.C.)
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| |
Collapse
|
26
|
Jaeschke H, Ramachandran A. Acetaminophen Hepatotoxicity: Paradigm for Understanding Mechanisms of Drug-Induced Liver Injury. ANNUAL REVIEW OF PATHOLOGY 2024; 19:453-478. [PMID: 38265880 PMCID: PMC11131139 DOI: 10.1146/annurev-pathmechdis-051122-094016] [Citation(s) in RCA: 47] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2024]
Abstract
Acetaminophen (APAP) overdose is the clinically most relevant drug hepatotoxicity in western countries, and, because of translational relevance of animal models, APAP is mechanistically the most studied drug. This review covers intracellular signaling events starting with drug metabolism and the central role of mitochondrial dysfunction involving oxidant stress and peroxynitrite. Mitochondria-derived endonucleases trigger nuclear DNA fragmentation, the point of no return for cell death. In addition, adaptive mechanisms that limit cell death are discussed including autophagy, mitochondrial morphology changes, and biogenesis. Extensive evidence supports oncotic necrosis as the mode of cell death; however, a partial overlap with signaling events of apoptosis, ferroptosis, and pyroptosis is the basis for controversial discussions. Furthermore, an update on sterile inflammation in injury and repair with activation of Kupffer cells, monocyte-derived macrophages, and neutrophils is provided. Understanding these mechanisms of cell death led to discovery of N-acetylcysteine and recently fomepizole as effective antidotes against APAP toxicity.
Collapse
Affiliation(s)
- Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA; ,
| | - Anup Ramachandran
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA; ,
| |
Collapse
|
27
|
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]
|
28
|
Rani J, Dhull SB, Rose PK, Kidwai MK. Drug-induced liver injury and anti-hepatotoxic effect of herbal compounds: a metabolic mechanism perspective. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 122:155142. [PMID: 37913641 DOI: 10.1016/j.phymed.2023.155142] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 09/27/2023] [Accepted: 10/10/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND Drug-induced liver injury (DILI) is the most challenging and thought-provoking liver problem for hepatologists owing to unregulated medication usage in medical practices, nutritional supplements, and botanicals. Due to underreporting, analysis, and identification issues, clinically evaluated medication hepatotoxicity is prevalent yet hard to quantify. PURPOSE This review's primary objective is to thoroughly compare pharmaceutical drugs and herbal compounds that have undergone clinical trials, focusing on their metabolic mechanisms contributing to the onset of liver illnesses and their hepatoprotective effects. METHODS The data was gathered from several online sources, such as PubMed, Scopus, Google Scholar, and Web of Science, using appropriate keywords. RESULTS The prevalence of conventional and herbal medicine is rising. A comprehensive understanding of the metabolic mechanism is necessary to mitigate the hepatotoxicity induced by drugs and facilitate the incorporation or substitution of herbal medicine instead of pharmaceuticals. Moreover, pre-clinical pharmacological research has the potential to facilitate the development of natural products as therapeutic agents, displaying promising possibilities for their eventual clinical implementation. CONCLUSIONS Acetaminophen, isoniazid, rifampicin, diclofenac, and pyrogallol have been identified as the most often reported synthetic drugs that produce hepatotoxicity by oxidative stress, inflammation, apoptosis, and fibrosis during the last several decades. Due to their ability to downregulate many factors (such as cytokines) and activate several enzyme/enzyme systems, herbal substances (such as Gingko biloba extract, curcumin, resveratrol, and silymarin) provide superior protection against harmful mechanisms which induce hepatotoxicity with fewer adverse effects than their synthetic counterparts.
Collapse
Affiliation(s)
- Jyoti Rani
- Department of Botany, Chaudhary Devi Lal University, Sirsa 125055, Haryana, India
| | - Sanju Bala Dhull
- Department of Food Science and Technology, Chaudhary Devi Lal University, Sirsa 125055, Haryana, India.
| | - Pawan Kumar Rose
- Department of Energy and Environmental Sciences, Chaudhary Devi Lal University, Sirsa 125055, Haryana, India.
| | - Mohd Kashif Kidwai
- Department of Energy and Environmental Sciences, Chaudhary Devi Lal University, Sirsa 125055, Haryana, India
| |
Collapse
|
29
|
Kim DM, Lee JH, Pan Q, Han HW, Shen Z, Eshghjoo S, Wu CS, Yang W, Noh JY, Threadgill DW, Guo S, Wright G, Alaniz R, Sun Y. Nutrient-sensing growth hormone secretagogue receptor in macrophage programming and meta-inflammation. Mol Metab 2024; 79:101852. [PMID: 38092245 PMCID: PMC10772824 DOI: 10.1016/j.molmet.2023.101852] [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: 10/11/2023] [Revised: 12/03/2023] [Accepted: 12/08/2023] [Indexed: 12/20/2023] Open
Abstract
OBJECTIVE Obesity-associated chronic inflammation, aka meta-inflammation, is a key pathogenic driver for obesity-associated comorbidity. Growth hormone secretagogue receptor (GHSR) is known to mediate the effects of nutrient-sensing hormone ghrelin in food intake and fat deposition. We previously reported that global Ghsr ablation protects against diet-induced inflammation and insulin resistance, but the site(s) of action and mechanism are unknown. Macrophages are key drivers of meta-inflammation. To unravel the role of GHSR in macrophages, we generated myeloid-specific Ghsr knockout mice (LysM-Cre;Ghsrf/f). METHODS LysM-Cre;Ghsrf/f and control Ghsrf/f mice were subjected to 5 months of high-fat diet (HFD) feeding to induce obesity. In vivo, metabolic profiling of food intake, physical activity, and energy expenditure, as well as glucose and insulin tolerance tests (GTT and ITT) were performed. At termination, peritoneal macrophages (PMs), epididymal white adipose tissue (eWAT), and liver were analyzed by flow cytometry and histology. For ex vivo studies, bone marrow-derived macrophages (BMDMs) were generated from the mice and treated with palmitic acid (PA) or lipopolysaccharide (LPS). For in vitro studies, macrophage RAW264.7 cells with Ghsr overexpression or Insulin receptor substrate 2 (Irs2) knockdown were studied. RESULTS We found that Ghsr expression in PMs was increased under HFD feeding. In vivo, HFD-fed LysM-Cre;Ghsrf/f mice exhibited significantly attenuated systemic inflammation and insulin resistance without affecting food intake or body weight. Tissue analysis showed that HFD-fed LysM-Cre;Ghsrf/f mice have significantly decreased monocyte/macrophage infiltration, pro-inflammatory activation, and lipid accumulation, showing elevated lipid-associated macrophages (LAMs) in eWAT and liver. Ex vivo, Ghsr-deficient macrophages protected against PA- or LPS-induced pro-inflammatory polarization, showing reduced glycolysis, increased fatty acid oxidation, and decreased NF-κB nuclear translocation. At molecular level, GHSR metabolically programs macrophage polarization through PKA-CREB-IRS2-AKT2 signaling pathway. CONCLUSIONS These novel results demonstrate that macrophage GHSR plays a key role in the pathogenesis of meta-inflammation, and macrophage GHSR promotes macrophage infiltration and induces pro-inflammatory polarization. These exciting findings suggest that GHSR may serve as a novel immunotherapeutic target for the treatment of obesity and its associated comorbidity.
Collapse
Affiliation(s)
- Da Mi Kim
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Jong Han Lee
- Department of Marine Bioindustry, Hanseo University, Seosan 31962, South Korea; USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College Medicine, Houston, TX 77030, USA
| | - Quan Pan
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Hye Won Han
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Zheng Shen
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Sahar Eshghjoo
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77807, USA; Agilent technologies, Aanta Clara, CA 95051, USA
| | - Chia-Shan Wu
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College Medicine, Houston, TX 77030, USA
| | - Wanbao Yang
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Ji Yeon Noh
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - David W Threadgill
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; Texas A&M Institute for Genome Sciences and Society, Department of Cell Biology and Genetics, Texas A&M University, College Station, TX 77843, USA
| | - Shaodong Guo
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Gus Wright
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX 77843, USA
| | - Robert Alaniz
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77807, USA; Tlaloc Therapeutics Inc., College Station, TX 77845, USA
| | - Yuxiang Sun
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College Medicine, Houston, TX 77030, USA.
| |
Collapse
|
30
|
Tian WS, Zhao J, Kim MK, Tae HJ, Kim IS, Ahn D, Hwang HP, Mao MX, Park BY. Veronica persica ameliorates acetaminophen-induced murine hepatotoxicity via attenuating oxidative stress and inflammation. Biomed Pharmacother 2023; 169:115898. [PMID: 37989029 DOI: 10.1016/j.biopha.2023.115898] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 11/23/2023] Open
Abstract
Excess acetaminophen (APAP) commonly causes severe acute liver injury (ALI), characterized by oxidative stress, pro-inflammatory responses, and hepatocyte damage. Veronica persica (VP) is a traditional medicine with antioxidant and anti-inflammatory properties. There is a paucity of information on its medicinal value, especially its potential mechanisms for alleviating ALI. This study aimed to clarify the ameliorative effects and intracellular mechanisms of VP on APAP-induced ALI via attenuating oxidative stress and inflammation. Mice were given VP for 7 days before exposure to APAP (300 mg/kg). The HPLC and radical scavenging assay found that VP contains 12 phenolic acids and 6 flavonoids, as well as show robust antioxidant capacity. In the APAP-induced ALI model, pre-treatment with VP significantly reduces APAP-induced hepatotoxicity by observing improved hepatocyte pathological injury and further confirmed by serum biochemical indicator. Also, the reduction of TUNEL-positive regions and the regulation of Bcl-2-associated X protein indicated that VP attenuates hepatocytotoxicity. Moreover, VP pre-intervention inhibits the formation of liver pro-inflammatory cytokines, the expression of inflammatory response genes, and increases in myeloperoxidase (MPO) in APAP-exposed mice. The elevated reduced glutathione (GSH) levels and decreased oxidative stress markers indicate that VP reduces APAP-promoted oxidative stress. Further study revealed that VP inhibited the phosphorylation of NF-κB/STAT3 cascade, blocked ERK and JNK phosphorylation, and activated AMP-activated protein kinase (AMPK). To sum up, this study demonstrated that VP exists hepatoprotective abilities on APAP-induced ALI, primarily by suppressing the phosphorylation of NF-κB/STAT3 cascade and ERK-JNK and inducing AMPK activation to alleviate oxidative stress and inflammation.
Collapse
Affiliation(s)
- Wei-Shun Tian
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan, People's Republic of China; College of Veterinary Medicine and Institute of Animal Transplantation, Jeonbuk National University, Iksan, Jeollabuk-do 54596, Republic of Korea
| | - Jing Zhao
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan, People's Republic of China
| | - Myung-Kon Kim
- Department of Food Science and Technology, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Hyun-Jin Tae
- College of Veterinary Medicine and Institute of Animal Transplantation, Jeonbuk National University, Iksan, Jeollabuk-do 54596, Republic of Korea
| | - In-Shik Kim
- College of Veterinary Medicine and Institute of Animal Transplantation, Jeonbuk National University, Iksan, Jeollabuk-do 54596, Republic of Korea
| | - Dongchoon Ahn
- College of Veterinary Medicine and Institute of Animal Transplantation, Jeonbuk National University, Iksan, Jeollabuk-do 54596, Republic of Korea
| | - Hong Pil Hwang
- Department of Surgery of Jeonbuk National University Medical School and Hospital, Jeonju 54896, Republic of Korea
| | - Ming-Xian Mao
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan, People's Republic of China
| | - Byung-Yong Park
- College of Veterinary Medicine and Institute of Animal Transplantation, Jeonbuk National University, Iksan, Jeollabuk-do 54596, Republic of Korea.
| |
Collapse
|
31
|
McGill MR, Curry SC. The Evolution of Circulating Biomarkers for Use in Acetaminophen/Paracetamol-Induced Liver Injury in Humans: A Scoping Review. LIVERS 2023; 3:569-596. [PMID: 38434489 PMCID: PMC10906739 DOI: 10.3390/livers3040039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/05/2024] Open
Abstract
Acetaminophen (APAP) is a widely used drug, but overdose can cause severe acute liver injury. The first reports of APAP hepatotoxicity in humans were published in 1966, shortly after the development of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) as the first biomarkers of liver injury as opposed to liver function. Thus, the field of liver injury biomarkers has evolved alongside the growth in APAP hepatotoxicity incidence. Numerous biomarkers have been proposed for use in the management of APAP overdose patients in the intervening years. Here, we comprehensively review the development of these markers from the 1960s to the present day and briefly discuss possible future directions.
Collapse
Affiliation(s)
- Mitchell R McGill
- Dept. of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR 72212, USA
- Dept. of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72212, USA
- Dept. of Pathology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72212, USA
| | - Steven C Curry
- Division of Clinical Data Analytics and Decision Support, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85006, USA
- Department of Medical Toxicology, Banner-University Medical Center Phoenix, Phoenix, AZ 85006, USA
| |
Collapse
|
32
|
Roth K, Strickland J, Pant A, Freeborn R, Kennedy R, Rockwell CE, Luyendyk JP, Copple BL. Interleukin-10 disrupts liver repair in acetaminophen-induced acute liver failure. Front Immunol 2023; 14:1303921. [PMID: 38094302 PMCID: PMC10716295 DOI: 10.3389/fimmu.2023.1303921] [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: 09/28/2023] [Accepted: 11/13/2023] [Indexed: 12/18/2023] Open
Abstract
Introduction Systemic levels of the anti-inflammatory cytokine interleukin 10 (IL-10) are highest in acetaminophen (APAP)-induced acute liver failure (ALF) patients with the poorest prognosis. The mechanistic basis for this counterintuitive finding is not known, as induction of IL-10 is hypothesized to temper the pathological effects of immune cell activation. Aberrant production of IL-10 after severe liver injury could conceivably interfere with the beneficial, pro-reparative actions of immune cells, such as monocytes. Methods To test this possibility, we determined whether IL-10 levels are dysregulated in mice with APAP-induced ALF and further evaluated whether aberrant production of IL-10 prevents monocyte recruitment and/or the resolution of necrotic lesions by these cells. Results Our studies demonstrate that in mice challenged with 300 mg/kg acetaminophen (APAP), a hepatotoxic dose of APAP that fails to produce ALF (i.e., APAP-induced acute liver injury; AALI), Ly6Chi monocytes were recruited to the liver and infiltrated the necrotic lesions by 48 hours coincident with the clearance of dead cell debris. At 72 hours, IL-10 was upregulated, culminating in the resolution of hepatic inflammation. By contrast, in mice treated with 600 mg/kg APAP, a dose that produces clinical features of ALF (i.e., APAP-induced ALF; AALF), IL-10 levels were markedly elevated by 24 hours. Early induction of IL-10 was associated with a reduction in the hepatic numbers of Ly6Chi monocytes resulting in the persistence of dead cell debris. Inhibition of IL-10 in AALF mice, beginning at 24 hours after APAP treatment, increased the hepatic numbers of monocytes which coincided with a reduction in the necrotic area. Moreover, pharmacologic elevation of systemic IL-10 levels in AALI mice reduced hepatic myeloid cell numbers and increased the area of necrosis. Discussion Collectively, these results indicate that during ALF, aberrant production of IL-10 disrupts the hepatic recruitment of monocytes, which prevents the clearance of dead cell debris. These are the first studies to document a mechanistic basis for the link between high IL-10 levels and poor outcome in patients with ALF.
Collapse
Affiliation(s)
- Katherine Roth
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Cell and Molecular Biology Program, Michigan State University, East Lansing, MI, United States
- College of Human Medicine, Michigan State University, East Lansing, MI, United States
| | - Jenna Strickland
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Cell and Molecular Biology Program, Michigan State University, East Lansing, MI, United States
- College of Human Medicine, Michigan State University, East Lansing, MI, United States
| | - Asmita Pant
- Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, United States
| | - Robert Freeborn
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
| | - Rebekah Kennedy
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
| | - Cheryl E. Rockwell
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Cell and Molecular Biology Program, Michigan State University, East Lansing, MI, United States
- College of Human Medicine, Michigan State University, East Lansing, MI, United States
| | - James P. Luyendyk
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, United States
| | - Bryan L. Copple
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Cell and Molecular Biology Program, Michigan State University, East Lansing, MI, United States
- College of Human Medicine, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
33
|
Zhao X, Yin F, Fu L, Ma Y, Ye L, Huang Y, Fan W, Gao W, Cai Y, Mou X. Garlic-derived exosome-like nanovesicles as a hepatoprotective agent alleviating acute liver failure by inhibiting CCR2/CCR5 signaling and inflammation. BIOMATERIALS ADVANCES 2023; 154:213592. [PMID: 37717364 DOI: 10.1016/j.bioadv.2023.213592] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/27/2023] [Accepted: 08/15/2023] [Indexed: 09/19/2023]
Abstract
Acute liver failure (ALF) is a life-threatening clinical syndrome mostly induced by viral infections or drug abuse. As a novel therapeutic adjuvant or delivery vehicle, plant-derived exosome-like nanovesicles (PELNVs) have been extensively studied in recent years. This study aimed to develop garlic-derived exosome-like nanovesicles (GaELNVs) in order to ameliorate liver injury induced by LPS/D-GalN in mice, inhibit inflammatory eruption and reduce inflammatory cells infiltration. The results showed that treatment with GaELNVs improved liver pathology and reduced the levels of soluble inflammatory mediators IL-6, IL-1β and TNF-α in the serum of ALF mice. GaELNVs reversed the upregulation of Cleaved Caspase-9, Cleaved Caspase-3, p53 and Bax expression and decreased Bcl2 activation caused by D-GalN/LPS, and inhibited NF-κB p65 expression and translocation to the nucleus. Meanwhile, treatment with GaELNVs resulted significant reduction in NLRP3 activation and Caspase-1 maturation, as well as decrease in the release of the inflammatory mediator IL-18. Additionally, an upregulation of the expression of proteins related to energy metabolism and autophagy occurrence including Foxo3a, Sirt1, and LC3-II was detected in the liver. Oral administration of GaELNVs also led to significant alteration in the expression of F4/80 and CD11b in the liver. Furthermore, the detection of chemokines in mouse liver tissue revealed that GaELNVs exhibited minimal reduction in the expression of CCL2, CCL3, CCL5 and CCL8. The decreased expression of CCR2 and CCR5 in the liver suggests that GaELNVs have the ability to decrease the recruitment of monocytes from the circulation to the liver. A reduction in the infiltration of F4/80loCD11bhi monocyte-derived macrophages into the liver was also observed. This study provides novel evidence that GaELNVs can ameliorate inflammatory eruptions and hinder the migration of circulating monocytes to the liver, as well as decrease macrophage infiltration by inhibiting CCR2/CCR5 signaling. Consequently, GaELNVs hold promise as a novel therapeutic agent for clinical management of liver disease.
Collapse
Affiliation(s)
- Xin Zhao
- General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China; College of Pharmacy, Hangzhou Medical College, Hangzhou 310059, China; Clinical Research Institute, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China
| | - Fang Yin
- Shanghai Engineering Research Center of Human Intestinal Microflora Function Development, Shanghai Tenth People's Hospital, Shanghai 200072, China
| | - Luoqin Fu
- Clinical Research Institute, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China
| | - Yingyu Ma
- Clinical Research Institute, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China
| | - Luyi Ye
- College of Pharmacy, Hangzhou Medical College, Hangzhou 310059, China
| | - Yilin Huang
- College of Pharmacy, Hangzhou Medical College, Hangzhou 310059, China
| | - Weijiao Fan
- Clinical Research Institute, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China
| | - Wenxue Gao
- Clinical Research Unit, Shanghai Tenth People's Hospital, Shanghai 200072, China.
| | - Yu Cai
- General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China; College of Pharmacy, Hangzhou Medical College, Hangzhou 310059, China; Clinical Research Institute, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China.
| | - Xiaozhou Mou
- General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China; College of Pharmacy, Hangzhou Medical College, Hangzhou 310059, China; Clinical Research Institute, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China.
| |
Collapse
|
34
|
Chou AH, Lee HC, Liao CC, Yu HP, Liu FC. ERK/NF-kB/COX-2 Signaling Pathway Plays a Key Role in Curcumin Protection against Acetaminophen-Induced Liver Injury. Life (Basel) 2023; 13:2150. [PMID: 38004290 PMCID: PMC10672507 DOI: 10.3390/life13112150] [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: 09/26/2023] [Revised: 10/23/2023] [Accepted: 10/30/2023] [Indexed: 11/26/2023] Open
Abstract
Recent experimental studies have highlighted the beneficial effects of curcumin on liver injury induced by acetaminophen (APAP). However, the specific molecular mechanisms underlying curcumin's hepatoprotective effects against APAP-induced liver injury remain to be fully elucidated. This study aimed to investigate the therapeutic effect of curcumin on APAP-induced liver injury using a mouse model. In the experiment, mice were subjected to an intraperitoneal hepatotoxic dose of APAP (300 mg/kg) to induce hepatotoxicity. After 30 min of APAP administration, the mice were treated with different concentrations of curcumin (0, 10, 25, or 50 mg/kg). After 16 h, mice with hepatotoxicity showed elevated levels of serum alanine transaminase (ALT), aspartate transaminase (AST), hepatic myeloperoxidase (MPO), TNF-α, and IL-6, and decreased levels of glutathione (GSH). Moreover, there was an increased infiltration of neutrophils and macrophages following intraperitoneal injection of APAP. However, curcumin-treated mice displayed a pronounced reduction in serum ALT, AST, hepatic MPO, TNF-α, and IL-6 levels, coupled with a notable elevation in GSH levels compared to the APAP-treated hepatotoxic mice. Moreover, curcumin treatment led to reduced infiltration of neutrophils and macrophages. Additionally, curcumin inhibited the phosphorylation of ERK and NF-kB proteins while reducing the expression of cyclooxygenase-2 (COX-2). These findings highlight the hepatoprotective potential of curcumin against APAP-induced liver injury through the suppression of the ERK, NF-kB, and COX-2 signaling pathways.
Collapse
Affiliation(s)
- An-Hsun Chou
- Department of Anesthesiology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333, Taiwan; (A.-H.C.); (H.-C.L.); (C.-C.L.); (H.-P.Y.)
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Hung-Chen Lee
- Department of Anesthesiology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333, Taiwan; (A.-H.C.); (H.-C.L.); (C.-C.L.); (H.-P.Y.)
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chia-Chih Liao
- Department of Anesthesiology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333, Taiwan; (A.-H.C.); (H.-C.L.); (C.-C.L.); (H.-P.Y.)
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Huang-Ping Yu
- Department of Anesthesiology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333, Taiwan; (A.-H.C.); (H.-C.L.); (C.-C.L.); (H.-P.Y.)
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Fu-Chao Liu
- Department of Anesthesiology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333, Taiwan; (A.-H.C.); (H.-C.L.); (C.-C.L.); (H.-P.Y.)
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| |
Collapse
|
35
|
Luo S, Zhao X, Jiang J, Deng B, Liu S, Xu H, Tan Q, Chen Y, Zhang Z, Pan X, Wan R, Chen X, Yao Y, Li J. Piezo1 specific deletion in macrophage protects the progression of liver fibrosis in mice. Theranostics 2023; 13:5418-5434. [PMID: 37908726 PMCID: PMC10614683 DOI: 10.7150/thno.86103] [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: 05/12/2023] [Accepted: 09/18/2023] [Indexed: 11/02/2023] Open
Abstract
Background and Aims: Liver fibrosis is the common pathological pathway of chronic liver diseases and its mechanisms of which have not been fully declared. Macrophages play essential roles in progression of liver fibrosis partially by sensing abnormal mechanical signals. The aim of the study is to investigate the functions of macrophage Piezo1, a mechano-sensitive ion channel, in liver fibrosis. Approach and Results: Immunofluorescence in human and murine fibrotic liver samples revealed that expression of macrophage Piezo1 was increased. Myeloid-specific Piezo1 knockout (Piezo1ΔLysM) attenuated liver fibrosis by decreased collagen deposition and epithelial-mesenchymal transition (EMT). In Piezo1ΔLysM mice, less inflammation during development of liver fibrosis was observed by lessened macrophage infiltration, decreased M1 polarization and expression of inflammatory cytokines. RNA-seq data showed macrophage Piezo1 regulated transcription of cathepsin S (CTSS). Piezo1ΔLysM inhibited expression and activity of CTSS in vitro and in vivo and regulated T cell activity. Furthermore, inhibition of CTSS reversed macrophage inflammatory response driven by Piezo1 activation and LPS. Macrophage Piezo1 activation promoted CTSS secretion due to increased activity of Ca2+-dependent calpain protease induced by Ca2+ influx to cleave lysosome-associated membrane protein-1 (LAMP1). Pharmacological inhibition of calpain activity partially blocked Piezo1 mediated CTSS secretion. Conclusions: Macrophage Piezo1 deficiency limits the progression of liver fibrosis by inhibited inflammatory response and decreased secretion of CTSS. These findings suggest that targeting Piezo1 channel may be a potential strategy for treating hepatic fibrosis.
Collapse
Affiliation(s)
- Shangfei Luo
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Innovation Research Center, Shandong University of Chinese Medicine, Jinan, 250307, China
| | - Xiaoduo Zhao
- Department of Pathology, the First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Jintao Jiang
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Bo Deng
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510260, China
| | - Silin Liu
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Honglin Xu
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Qiaorui Tan
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Yu'an Chen
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Ziyan Zhang
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Xianmei Pan
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Rentao Wan
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Xiaoting Chen
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Youfen Yao
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Jing Li
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Innovation Research Center, Shandong University of Chinese Medicine, Jinan, 250307, China
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, LS2 9JT, UK
| |
Collapse
|
36
|
Geng Y, Wang J, Serna-Salas SA, Villanueva AH, Buist-Homan M, Arrese M, Olinga P, Blokzijl H, Moshage H. Hepatic stellate cells induce an inflammatory phenotype in Kupffer cells via the release of extracellular vesicles. J Cell Physiol 2023; 238:2293-2303. [PMID: 37555553 DOI: 10.1002/jcp.31086] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/15/2023] [Accepted: 07/12/2023] [Indexed: 08/10/2023]
Abstract
Liver fibrosis is the response of the liver to chronic liver inflammation. The communication between the resident liver macrophages (Kupffer cells [KCs]) and hepatic stellate cells (HSCs) has been mainly viewed as one-directional: from KCs to HSCs with KCs promoting fibrogenesis. However, recent studies indicated that HSCs may function as a hub of intercellular communications. Therefore, the aim of the present study was to investigate the role of HSCs on the inflammatory phenotype of KCs. Primary rat HSCs and KCs were isolated from male Wistar rats. HSCs-derived conditioned medium (CM) was harvested from different time intervals (Day 0-2: CM-D2 and Day 5-7: CM-D7) during the activation of HSCs. Extracellular vesicles (EVs) were isolated from CM by ultracentrifugation and evaluated by nanoparticle tracking analysis and western blot analysis. M1 and M2 markers of inflammation were measured by quantitative PCR and macrophage function by assessing phagocytic capacity. CM-D2 significantly induced the inflammatory phenotype in KCs, but not CM-D7. Neither CM-D2 nor CM-D7 affected the phagocytosis of KCs. Importantly, the proinflammatory effect of HSCs-derived CM is mediated via EVs released from HSCs since EVs isolated from CM mimicked the effect of CM, whereas EV-depleted CM lost its ability to induce a proinflammatory phenotype in KCs. In addition, when the activation of HSCs was inhibited, HSCs produced less EVs. Furthermore, the proinflammatory effects of CM and EVs are related to activating Toll-like receptor 4 (TLR4) in KCs. In conclusion, HSCs at an early stage of activation induce a proinflammatory phenotype in KCs via the release of EVs. This effect is absent in CM derived from HSCs at a later stage of activation and is dependent on the activation of TLR4 signaling pathway.
Collapse
Affiliation(s)
- Yana Geng
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Junyu Wang
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Sandra Alejandra Serna-Salas
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Alejandra Hernández Villanueva
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Gastroenterology, Pontificia Universidad Católica de Chile, Santiago de Chile, Chile
| | - Manon Buist-Homan
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Marco Arrese
- Department of Gastroenterology, Pontificia Universidad Católica de Chile, Santiago de Chile, Chile
| | - Peter Olinga
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Hans Blokzijl
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Han Moshage
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
37
|
Yang W, Kim DM, Jiang W, Ai W, Pan Q, Rahman S, Cai JJ, Brashear WA, Sun Y, Guo S. Suppression of FOXO1 attenuates inflamm-aging and improves liver function during aging. Aging Cell 2023; 22:e13968. [PMID: 37602516 PMCID: PMC10577549 DOI: 10.1111/acel.13968] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/04/2023] [Indexed: 08/22/2023] Open
Abstract
The liver is a key metabolic organ that maintains whole-body nutrient homeostasis. Aging-induced liver function alterations contribute to systemic susceptibility to aging-related diseases. However, the molecular mechanisms of liver aging remain insufficiently understood. In this study, we performed bulk RNA-Seq and single-cell RNA-Seq analyses to investigate the underlying mechanisms of the aging-induced liver function changes. We found that liver inflammation, glucose intolerance, and liver fat deposition were aggravated in old mice. Aging significantly increased pro-inflammation in hepatic macrophages. Furthermore, we found that Kupffer cells (KCs) were the major driver to induce pro-inflammation in hepatic macrophages during aging. In KCs, aging significantly increased pro-inflammatory levels; in monocyte-derived macrophages (MDMs), aging had a limited effect on pro-inflammation but led to a functional quiescence in antigen presentation and phagosome process. In addition, we identified an aging-responsive KC-specific (ARKC) gene set that potentially mediates aging-induced pro-inflammation in KCs. Interestingly, FOXO1 activity was significantly increased in the liver of old mice. FOXO1 inhibition by AS1842856 significantly alleviated glucose intolerance, hepatic steatosis, and systemic inflammation in old mice. FOXO1 inhibition significantly attenuated aging-induced pro-inflammation in KCs partially through downregulation of ARKC genes. However, FOXO1 inhibition had a limited effect on aging-induced functional quiescence in MDMs. These results indicate that aging induces pro-inflammation in liver mainly through targeting KCs and FOXO1 is a key player in aging-induced pro-inflammation in KCs. Thus, FOXO1 could be a potential therapeutic target for the treatment of age-associated chronic diseases.
Collapse
Affiliation(s)
- Wanbao Yang
- Department of Nutrition, College of Agriculture and Life SciencesTexas A&M UniversityCollege StationTexasUSA
| | - Da Mi Kim
- Department of Nutrition, College of Agriculture and Life SciencesTexas A&M UniversityCollege StationTexasUSA
| | - Wen Jiang
- Department of Nutrition, College of Agriculture and Life SciencesTexas A&M UniversityCollege StationTexasUSA
| | - Weiqi Ai
- Department of Nutrition, College of Agriculture and Life SciencesTexas A&M UniversityCollege StationTexasUSA
| | - Quan Pan
- Department of Nutrition, College of Agriculture and Life SciencesTexas A&M UniversityCollege StationTexasUSA
| | - Shahina Rahman
- Department of StatisticsTexas A&M UniversityCollege StationTexasUSA
| | - James J. Cai
- Department of Veterinary Integrative BiosciencesTexas A&M UniversityCollege StationTexasUSA
| | - Wesley A. Brashear
- High Performance Research ComputingTexas A&M UniversityCollege StationTexasUSA
| | - Yuxiang Sun
- Department of Nutrition, College of Agriculture and Life SciencesTexas A&M UniversityCollege StationTexasUSA
| | - Shaodong Guo
- Department of Nutrition, College of Agriculture and Life SciencesTexas A&M UniversityCollege StationTexasUSA
| |
Collapse
|
38
|
Brown ZJ, Ruff SM, Pawlik TM. The effect of liver disease on hepatic microenvironment and implications for immune therapy. Front Pharmacol 2023; 14:1225821. [PMID: 37608898 PMCID: PMC10441240 DOI: 10.3389/fphar.2023.1225821] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 07/31/2023] [Indexed: 08/24/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver cancer and the fourth leading cause of cancer-related death worldwide. HCC often occurs in the setting of chronic liver disease or cirrhosis. Recent evidence has highlighted the importance of the immune microenvironment in the development and progression of HCC, as well as its role in the potential response to therapy. Liver disease such as viral hepatitis, alcohol induced liver disease, and non-alcoholic fatty liver disease is a major risk factor for the development of HCC and has been demonstrated to alter the immune microenvironment. Alterations in the immune microenvironment may markedly influence the response to different therapeutic strategies. As such, research has focused on understanding the complex relationship among tumor cells, immune cells, and the surrounding liver parenchyma to treat HCC more effectively. We herein review the immune microenvironment, as well as the relative effect of liver disease on the immune microenvironment. In addition, we review how changes in the immune microenvironment can lead to therapeutic resistance, as well as highlight future strategies aimed at developing the next-generation of therapies for HCC.
Collapse
Affiliation(s)
- Zachary J. Brown
- Department of Surgery, New York University Long Island School of Medicine, Mineola, NY, United States
| | - Samantha M. Ruff
- James Comprehensive Cancer Center, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Timothy M. Pawlik
- James Comprehensive Cancer Center, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
39
|
Peng B, Li H, Liu K, Zhang P, Zhuang Q, Li J, Yang M, Cheng K, Ming Y. Intrahepatic macrophage reprogramming associated with lipid metabolism in hepatitis B virus-related acute-on-chronic liver failure. J Transl Med 2023; 21:419. [PMID: 37380987 DOI: 10.1186/s12967-023-04294-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 06/22/2023] [Indexed: 06/30/2023] Open
Abstract
BACKGROUND Acute-on-chronic liver failure (ACLF) is a severe syndrome with high short-term mortality, but the pathophysiology still remains largely unknown. Immune dysregulation and metabolic disorders contribute to the progression of ACLF, but the crosstalk between immunity and metabolism during ACLF is less understood. This study aims to depict the immune microenvironment in the liver during ACLF, and explore the role of lipid metabolic disorder on immunity. METHODS Single-cell RNA-sequencing (scRNA-seq) was performed using the liver non-parenchymal cells (NPCs) and peripheral blood mononuclear cells (PBMCs) from healthy controls, cirrhosis patients and ACLF patients. A series of inflammation-related cytokines and chemokines were detected using liver and plasma samples. The lipid metabolomics targeted free fatty acids (FFAs) in the liver was also detected. RESULTS The scRNA-seq analysis of liver NPCs showed a significant increase of monocytes/macrophages (Mono/Mac) infiltration in ACLF livers, whereas the resident Kupffer cells (KCs) were exhausted. A characterized TREM2+ Mono/Mac subpopulation was identified in ACLF, and showed immunosuppressive function. Combined with the scRNA-seq data from PBMCs, the pseudotime analysis revealed that the TREM2+ Mono/Mac were differentiated from the peripheral monocytes and correlated with lipid metabolism-related genes including APOE, APOC1, FABP5 and TREM2. The targeted lipid metabolomics proved the accumulation of unsaturated FFAs associated with α-linolenic acid (α-LA) and α-LA metabolism and beta oxidation of very long chain fatty acids in the ACLF livers, indicating that unsaturated FFAs might promote the differentiation of TREM2+ Mono/Mac during ACLF. CONCLUSIONS The reprogramming of macrophages was found in the liver during ACLF. The immunosuppressive TREM2+ macrophages were enriched in the ACLF liver and contributed to the immunosuppressive hepatic microenvironment. The accumulation of unsaturated FFAs in the ACLF liver promoted the reprogramming of the macrophages. It might be a potential target to improve the immune deficiency of ACLF patients through regulating lipid metabolism.
Collapse
Affiliation(s)
- Bo Peng
- Transplantation Center, The Third Xiangya Hospital, Central South University, Hunan, 410013, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Hao Li
- Transplantation Center, The Third Xiangya Hospital, Central South University, Hunan, 410013, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Kai Liu
- Transplantation Center, The Third Xiangya Hospital, Central South University, Hunan, 410013, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Pengpeng Zhang
- Transplantation Center, The Third Xiangya Hospital, Central South University, Hunan, 410013, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Quan Zhuang
- Transplantation Center, The Third Xiangya Hospital, Central South University, Hunan, 410013, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Junhui Li
- Transplantation Center, The Third Xiangya Hospital, Central South University, Hunan, 410013, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Min Yang
- Transplantation Center, The Third Xiangya Hospital, Central South University, Hunan, 410013, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Ke Cheng
- Transplantation Center, The Third Xiangya Hospital, Central South University, Hunan, 410013, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Yingzi Ming
- Transplantation Center, The Third Xiangya Hospital, Central South University, Hunan, 410013, Changsha, China.
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China.
| |
Collapse
|
40
|
Rani R, Gandhi CR. Stellate cell in hepatic inflammation and acute injury. J Cell Physiol 2023; 238:1226-1236. [PMID: 37120832 DOI: 10.1002/jcp.31029] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/05/2023] [Accepted: 04/07/2023] [Indexed: 05/02/2023]
Abstract
The perisinusoidal hepatic stellate cells (HSCs) have been investigated extensively for their role as the major fibrogenic cells during chronic liver injury. HSCs also produce numerous cytokines, chemokines, and growth mediators, and express cell adhesion molecules constitutively and in response to stimulants such as endotoxin (lipopolysaccharide). With this property and by interacting with resident and recruited immune and inflammatory cells, HSCs regulate hepatic immune homeostasis, inflammation, and acute injury. Indeed, experiments with HSC-depleted animal models and cocultures have provided evidence for the prominent role of HSCs in the initiation and progression of inflammation and acute liver damage due to various toxic agents. Thus HSCs and/or mediators derived thereof during acute liver damage may be considered as potential therapeutic targets.
Collapse
Affiliation(s)
- Richa Rani
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Research & Development, Cincinnati Veterans Administration Medical Center, Cincinnati, Ohio, USA
| | - Chandrashekhar R Gandhi
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Research & Development, Cincinnati Veterans Administration Medical Center, Cincinnati, Ohio, USA
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
41
|
Stern S, Wang H, Sadrieh N. Microphysiological Models for Mechanistic-Based Prediction of Idiosyncratic DILI. Cells 2023; 12:1476. [PMID: 37296597 PMCID: PMC10253021 DOI: 10.3390/cells12111476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/18/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Drug-induced liver injury (DILI) is a major contributor to high attrition rates among candidate and market drugs and a key regulatory, industry, and global health concern. While acute and dose-dependent DILI, namely, intrinsic DILI, is predictable and often reproducible in preclinical models, the nature of idiosyncratic DILI (iDILI) limits its mechanistic understanding due to the complex disease pathogenesis, and recapitulation using in vitro and in vivo models is extremely challenging. However, hepatic inflammation is a key feature of iDILI primarily orchestrated by the innate and adaptive immune system. This review summarizes the in vitro co-culture models that exploit the role of the immune system to investigate iDILI. Particularly, this review focuses on advancements in human-based 3D multicellular models attempting to supplement in vivo models that often lack predictability and display interspecies variations. Exploiting the immune-mediated mechanisms of iDILI, the inclusion of non-parenchymal cells in these hepatoxicity models, namely, Kupffer cells, stellate cells, dendritic cells, and liver sinusoidal endothelial cells, introduces heterotypic cell-cell interactions and mimics the hepatic microenvironment. Additionally, drugs recalled from the market in the US between 1996-2010 that were studies in these various models highlight the necessity for further harmonization and comparison of model characteristics. Challenges regarding disease-related endpoints, mimicking 3D architecture with different cell-cell contact, cell source, and the underlying multi-cellular and multi-stage mechanisms are described. It is our belief that progressing our understanding of the underlying pathogenesis of iDILI will provide mechanistic clues and a method for drug safety screening to better predict liver injury in clinical trials and post-marketing.
Collapse
Affiliation(s)
- Sydney Stern
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, MD 21201, USA;
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, MD 21201, USA;
| | - Nakissa Sadrieh
- Office of New Drugs, Center of Drug Evaluation and Research, FDA, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA
| |
Collapse
|
42
|
Park SJ, Garcia Diaz J, Um E, Hahn YS. Major roles of kupffer cells and macrophages in NAFLD development. Front Endocrinol (Lausanne) 2023; 14:1150118. [PMID: 37274349 PMCID: PMC10235620 DOI: 10.3389/fendo.2023.1150118] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 05/09/2023] [Indexed: 06/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is an important public health problem with growing numbers of NAFLD patients worldwide. Pathological conditions are different in each stage of NAFLD due to various factors. Preclinical and clinical studies provide evidence for a crucial role of immune cells in NAFLD progression. Liver-resident macrophages, kupffer cells (KCs), and monocytes-derived macrophages are the key cell types involved in the progression of NAFLD, non-alcoholic steatohepatitis (NASH), and hepatocellular carcinoma (HCC). Their unique polarization contributes to the progression of NAFLD. KCs are phagocytes with self-renewal abilities and play a role in regulating and maintaining homeostasis. Upon liver damage, KCs are activated and colonized at the site of the damaged tissue. The secretion of inflammatory cytokines and chemokines by KCs play a pivotal role in initiating NAFLD pathogenesis. This review briefly describes the role of immune cells in the immune system in NAFLD, and focuses on the pathological role and molecular pathways of KCs and recruited macrophages. In addition, the relationship between macrophages and insulin resistance is described. Finally, the latest therapeutics that target KCs and macrophages are summarized for the prevention and treatment of NAFLD.
Collapse
Affiliation(s)
- Soo-Jeung Park
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
| | - Josefina Garcia Diaz
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, United States
| | - Eugene Um
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
| | - Young S. Hahn
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
43
|
Nguyen NT, Umbaugh DS, Smith S, Adelusi OB, Sanchez-Guerrero G, Ramachandran A, Jaeschke H. Dose-dependent pleiotropic role of neutrophils during acetaminophen-induced liver injury in male and female mice. Arch Toxicol 2023; 97:1397-1412. [PMID: 36928416 PMCID: PMC10680445 DOI: 10.1007/s00204-023-03478-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 03/02/2023] [Indexed: 03/18/2023]
Abstract
Acetaminophen (APAP) overdose is the leading cause of acute liver failure in western countries. APAP can cause extensive hepatocellular necrosis, which triggers an inflammatory response involving neutrophil and monocyte recruitment. Particularly the role of neutrophils in the injury mechanism of APAP hepatotoxicity has been highly controversial. Thus, the objective of the current study was to assess whether a potential contribution of neutrophils was dependent on the APAP dose and the sex of the animals. Male and female C57BL/6 J mice were treated with 300 or 600 mg/kg APAP and the injury and inflammatory cell recruitment was evaluated between 6 and 48 h. In both male and female mice, ALT plasma levels and the areas of necrosis peaked at 12-24 h after both doses with more severe injury at the higher dose. In addition, Ly6g-positive neutrophils started to accumulate in the liver at 6 h and peaked at 6-12 h after 300 mg/kg and 12-24 h after 600 mg/kg for both sexes; however, the absolute numbers of hepatic neutrophils in the liver were significantly higher after the 600 mg/kg dose. Neutrophil infiltration correlated with mRNA levels of the neutrophil chemoattractant Cxcl2 in the liver. Treating mice with an anti-Cxcl2 antibody at 2 h after APAP significantly reduced neutrophil accumulation at 24 h after both doses and in both sexes. However, the injury was significantly reduced only after the high overdose. Thus, neutrophils, recruited through Cxcl2, have no effect on APAP-induced liver injury after 300 mg/kg but aggravate the injury only after severe overdoses.
Collapse
Affiliation(s)
- Nga T Nguyen
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd, MS 1018, Kansas City, Kansas, 66160, USA
| | - David S Umbaugh
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd, MS 1018, Kansas City, Kansas, 66160, USA
| | - Sawyer Smith
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd, MS 1018, Kansas City, Kansas, 66160, USA
| | - Olamide B Adelusi
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd, MS 1018, Kansas City, Kansas, 66160, USA
| | - Giselle Sanchez-Guerrero
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd, MS 1018, Kansas City, Kansas, 66160, USA
| | - Anup Ramachandran
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd, MS 1018, Kansas City, Kansas, 66160, USA
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd, MS 1018, Kansas City, Kansas, 66160, USA.
| |
Collapse
|
44
|
Ito Y, Hosono K, Amano H. Responses of hepatic sinusoidal cells to liver ischemia–reperfusion injury. Front Cell Dev Biol 2023; 11:1171317. [PMID: 37082623 PMCID: PMC10112669 DOI: 10.3389/fcell.2023.1171317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 03/27/2023] [Indexed: 04/07/2023] Open
Abstract
The liver displays a remarkable regenerative capacity in response to acute liver injury. In addition to the proliferation of hepatocytes during liver regeneration, non-parenchymal cells, including liver macrophages, liver sinusoidal endothelial cells (LSECs), and hepatic stellate cells (HSCs) play critical roles in liver repair and regeneration. Liver ischemia–reperfusion injury (IRI) is a major cause of increased liver damage during liver resection, transplantation, and trauma. Impaired liver repair increases postoperative morbidity and mortality of patients who underwent liver surgery. Successful liver repair and regeneration after liver IRI requires coordinated interplay and synergic actions between hepatic resident cells and recruited cell components. However, the underlying mechanisms of liver repair after liver IRI are not well understood. Recent technological advances have revealed the heterogeneity of each liver cell component in the steady state and diseased livers. In this review, we describe the progress in the biology of liver non-parenchymal cells obtained from novel technological advances. We address the functional role of each cell component in response to liver IRI and the interactions between diverse immune repertoires and non-hematopoietic cell populations during the course of liver repair after liver IRI. We also discuss how these findings can help in the design of novel therapeutic approaches. Growing insights into the cellular interactions during liver IRI would enhance the pathology of liver IRI understanding comprehensively and further develop the strategies for improvement of liver repair.
Collapse
|
45
|
Cathelicidin promotes liver repair after acetaminophen-induced liver injury in mice. JHEP Rep 2023; 5:100687. [PMID: 36923240 PMCID: PMC10009536 DOI: 10.1016/j.jhepr.2023.100687] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 01/09/2023] [Accepted: 01/12/2023] [Indexed: 02/01/2023] Open
Abstract
Background & Aims Acetaminophen (APAP)-induced acute liver injury (AILI) is a leading cause of acute liver failure (ALF). N-acetylcysteine (NAC) is only effective within 24 h after APAP intoxication, raising an urgent need for alternative approaches to treat this disease. This study aimed to test whether cathelicidin (Camp), which is a protective factor in chronic liver diseases, protects mice against APAP-induced liver injury and ALF. Methods A clinically relevant AILI model and an APAP-induced ALF model were generated in mice. Genetic and pharmacological approaches were used to interfere with the levels of cathelicidin in vivo. Results An increase in hepatic pro-CRAMP/CRAMP (the precursor and mature forms of mouse cathelicidin) was observed in APAP-intoxicated mice. Upregulated cathelicidin was derived from liver-infiltrating neutrophils. Compared with wild-type littermates, Camp knockout had no effect on hepatic injury but dampened hepatic repair in AILI and reduced survival in APAP-induced ALF. CRAMP administration reversed impaired liver recovery observed in APAP-challenged Camp knockout mice. Delayed CRAMP, CRAMP(1-39) (the extended form of CRAMP), or LL-37 (the mature form of human cathelicidin) treatment exhibited a therapeutic benefit for AILI. Co-treatment of cathelicidin and NAC in AILI displayed a stronger hepatoprotective effect than NAC alone. A similar additive effect of CRAMP(1-39)/LL-37 and NAC was observed in APAP-induced ALF. The pro-reparative role of cathelicidin in the APAP-damaged liver was attributed to an accelerated resolution of inflammation at the onset of liver repair, possibly through enhanced neutrophil phagocytosis of necrotic cell debris in an autocrine manner. Conclusions Cathelicidin reduces APAP-induced liver injury and ALF in mice by promoting liver recovery via facilitating inflammation resolution, suggesting a therapeutic potential for late-presenting patients with AILI with or without ALF. Impact and implications Acetaminophen-induced acute liver injury is a leading cause of acute liver failure. The efficacy of N-acetylcysteine, the only clinically approved drug against acetaminophen-induced acute liver injury, is significantly reduced for late-presenting patients. We found that cathelicidin exhibits a great therapeutic potential in mice with acetaminophen-induced liver injury or acute liver failure, which makes up for the limitation of N-acetylcysteine therapy by specifically promoting liver repair after acetaminophen intoxication. The pro-reparative role of cathelicidin, as a key effector molecule of neutrophils, in the APAP-injured liver is attributed to an accelerated resolution of inflammation at the onset of liver repair, possibly through enhanced phagocytic function of neutrophils in an autocrine manner.
Collapse
Key Words
- AILI, acetaminophen-induced acute liver injury
- ALF, acute liver failure
- ALT, alanine aminotransferase
- APAP, acetaminophen
- Acetaminophen
- CRAMP, cathelicidin-related antimicrobial peptide
- CYP2E1, cytochrome P450 2E1
- Cathelicidin
- EGF, endothelial growth factor
- FPR2/ALX, formyl peptide receptor type 2/lipoxin A4 receptor
- GSH, glutathione
- Inflammation resolution
- JNK, c-Jun N-terminal kinase
- KO, knockout
- Liver repair
- Mac-1, macrophage-1 antigen
- NAC, N-acetylcysteine
- NAPQI, N-acetyl-p-benzoquinone imine
- NPC, non-parenchymal cell
- Neutrophils
- Phagocytosis
- ROS, reactive oxygen species
- TLR4, Toll-like receptor 4
- WT, wild-type
- hCAP18, human cationic antimicrobial protein
- α-SMA, alpha-smooth muscle actin
Collapse
|
46
|
Xu L, Wang H. A dual role of inflammation in acetaminophen-induced liver injury. LIVER RESEARCH 2023; 7:9-15. [PMID: 39959696 PMCID: PMC11791818 DOI: 10.1016/j.livres.2023.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/14/2023] [Accepted: 03/05/2023] [Indexed: 03/12/2023]
Abstract
In many affluent nations, acetaminophen (APAP) overdose is the leading cause of drug-induced acute liver failure. The process of APAP-induced liver injury (AILI) is intimately tied to inflammation, including hepatocyte necrosis-caused initiation of inflammation, inflammation amplification that exacerbates liver injury, and the resolution of inflammation that triggers liver regeneration and repair. Excessive APAP metabolism in the liver eventually leads to hepatocyte necrosis and inflammation. Innate immune cells, such as neutrophils, eosinophils, monocytes, and gammadelta T cells, are recruited into the injured liver and release various cytokines. These immune cells and cytokines have been found to serve two purposes in AILI. In this review, we highlighted the dual role of inflammation, including inflammatory cytokines and inflammatory immune cells in AILI, and discussed possible explanations for contradictory findings.
Collapse
Affiliation(s)
- Long Xu
- School of Basic Medical Science, Anhui Medical University, Hefei, Anhui, China
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
47
|
Zhang H, Ni M, Wang H, Zhang J, Jin D, Busuttil RW, Kupiec-Weglinski JW, Li W, Wang X, Zhai Y. Gsk3β regulates the resolution of liver ischemia/reperfusion injury via MerTK. JCI Insight 2023; 8:e151819. [PMID: 36422999 PMCID: PMC9870084 DOI: 10.1172/jci.insight.151819] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 11/21/2022] [Indexed: 11/27/2022] Open
Abstract
Although glycogen synthase kinase β (Gsk3β) has been shown to regulate tissue inflammation, whether and how it regulates inflammation resolution versus inflammation activation is unclear. In a murine liver, partial warm ischemia/reperfusion injury (IRI) model, we found that Gsk3β inhibitory phosphorylation increased at both the early-activation and late-resolution stages of the disease. Myeloid Gsk3β deficiency not only alleviated liver injuries, it also facilitated the restoration of liver homeostasis. Depletion of Kupffer cells prior to the onset of liver ischemia diminished the differences between the WT and Gsk3β-KO mice in the activation of liver IRI. However, the resolution of liver IRI remained accelerated in Gsk3β-KO mice. In CD11b-DTR mice, Gsk3β-deficient BM-derived macrophages (BMMs) facilitated the resolution of liver IRI as compared with WT cells. Furthermore, Gsk3β deficiency promoted the reparative phenotype differentiation in vivo in liver-infiltrating macrophages and in vitro in BMMs. Gsk3 pharmacological inhibition promoted the resolution of liver IRI in WT, but not myeloid MerTK-deficient, mice. Thus, Gsk3β regulates liver IRI at both activation and resolution stages of the disease. Gsk3 inactivation enhances the proresolving function of liver-infiltrating macrophages in an MerTK-dependent manner.
Collapse
Affiliation(s)
- Hanwen Zhang
- Dumont-UCLA Transplant Center, Department of Surgery, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
- Department of Hepatobiliary-Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Ming Ni
- Dumont-UCLA Transplant Center, Department of Surgery, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
- Hepatobiliary Center, Key Laboratory of Liver Transplantation of Chinese Academy of Medical Sciences, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Han Wang
- Dumont-UCLA Transplant Center, Department of Surgery, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
- Hepatobiliary Center, Key Laboratory of Liver Transplantation of Chinese Academy of Medical Sciences, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Zhang
- Dumont-UCLA Transplant Center, Department of Surgery, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Dan Jin
- Dumont-UCLA Transplant Center, Department of Surgery, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ronald W. Busuttil
- Dumont-UCLA Transplant Center, Department of Surgery, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Jerzy W. Kupiec-Weglinski
- Dumont-UCLA Transplant Center, Department of Surgery, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Wei Li
- Department of Hepatobiliary-Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xuehao Wang
- Hepatobiliary Center, Key Laboratory of Liver Transplantation of Chinese Academy of Medical Sciences, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuan Zhai
- Dumont-UCLA Transplant Center, Department of Surgery, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
- Transplant Surgery, College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
48
|
Liu J, Zhang L, Xu F, Meng S, Li H, Song Y. Polystyrene Microplastics Postpone APAP-Induced Liver Injury through Impeding Macrophage Polarization. TOXICS 2022; 10:792. [PMID: 36548625 PMCID: PMC9781384 DOI: 10.3390/toxics10120792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/11/2022] [Accepted: 12/13/2022] [Indexed: 06/17/2023]
Abstract
Polystyrene microplastics (PS MPs) are micrometer-scale items degraded from plastics and have been detected in various organisms. PS MPs have been identified as causing cognitive, cardiac, intestinal, and hepatic damage. However, their role in liver regeneration under drug-induced liver injury remains unknown. Thus, the current study aims to evaluate the impact of PS MPs on liver repair during APAP hepatotoxicity. PS MPs pretreatment exacerbates mice mortality and hepatocyte apoptosis, suppresses hepatic cell proliferation, and disturbs the inflammatory response in the APAP-induced damage model. Further mechanism exploration uncovers that prior PS MPs administration is sufficient to recruit neutrophils and macrophages, which are necessary for tissue recovery in the acute liver injury model. However, the polarization capacity of macrophages to anti-inflammatory sub-type is significantly delayed in PS MPs plus APAP group compared to the single APAP group, which is the leading cause of tissue repair suppression. Overall, the current study supports a new insight to realize the toxicity of PS MPs in acute liver injury, which should be considered in health risk assessment.
Collapse
Affiliation(s)
- Jing Liu
- College of Eco-Environmental Engineering, Guizhou Minzu University, Guiyang 550025, China
- The Institute of Karst Wetland Ecology, Guizhou Minzu University, Guiyang 550025, China
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Lecong Zhang
- College of Eco-Environmental Engineering, Guizhou Minzu University, Guiyang 550025, China
| | - Fang Xu
- College of Eco-Environmental Engineering, Guizhou Minzu University, Guiyang 550025, China
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Songyan Meng
- College of Eco-Environmental Engineering, Guizhou Minzu University, Guiyang 550025, China
| | - Haitian Li
- College of Eco-Environmental Engineering, Guizhou Minzu University, Guiyang 550025, China
| | - Yang Song
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| |
Collapse
|
49
|
Woolbright BL, Nguyen NT, McGill MR, Sharpe MR, Curry SC, Jaeschke H. Generation of pro-and anti-inflammatory mediators after acetaminophen overdose in surviving and non-surviving patients. Toxicol Lett 2022; 367:59-66. [PMID: 35905941 PMCID: PMC9849076 DOI: 10.1016/j.toxlet.2022.07.813] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 07/17/2022] [Accepted: 07/25/2022] [Indexed: 01/21/2023]
Abstract
Acetaminophen (APAP) overdose causes liver injury in animals and humans. Although well-studied in animals, limited longitudinal data exist on cytokine release after APAP overdose in patients. The purpose of this study was to quantify concentrations of cytokines in APAP overdose patients to determine if early cytokine or complement measurements can distinguish between surviving and non-surviving patients. Plasma was obtained from healthy controls, APAP overdose patients with no increase in liver transaminases, and surviving and non-surviving APAP overdose patients with severe liver injury. Interleukin-10 (IL-10), and CC chemokine ligand-2 (CCL2, MCP-1) were substantially elevated in surviving and non-surviving patients, whereas IL-6 and CXC chemokine ligand-8 (CXCL8, IL-8) had early elevations in a subset of patients only with liver injury. Day 1 IL-10 and IL-6 levels, and Day 2 CCL2, levels correlated positively with survival. There was no significant increase in IL-1α, IL-1β or TNF-α in any patient during the first week after APAP. Monitoring cytokines such as CCL2 may be a good indicator of patient prognosis; furthermore, these data indicate the inflammatory response after APAP overdose in patients is not mediated by a second phase of inflammation driven by the inflammasome.
Collapse
Affiliation(s)
| | - Nga T Nguyen
- Department of Pharmacology, Toxicology & Therapeutics, USA
| | | | - Matthew R Sharpe
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Steven C Curry
- Department of Medical Toxicology, Banner Good Samaritan Medical Center, Phoenix, AZ, USA; Department of Medicine, and Center for Toxicology and Pharmacology Education and Research, University of Arizona College of Medicine, Phoenix, AZ, USA
| | | |
Collapse
|
50
|
Unraveling the effect of intra- and intercellular processes on acetaminophen-induced liver injury. NPJ Syst Biol Appl 2022; 8:27. [PMID: 35933513 PMCID: PMC9357019 DOI: 10.1038/s41540-022-00238-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 07/20/2022] [Indexed: 11/09/2022] Open
Abstract
In high dosages, acetaminophen (APAP) can cause severe liver damage, but susceptibility to liver failure varies across individuals and is influenced by factors such as health status. Because APAP-induced liver injury and recovery is regulated by an intricate system of intra- and extracellular molecular signaling, we here aim to quantify the importance of specific modules in determining the outcome after an APAP insult and of potential targets for therapies that mitigate adversity. For this purpose, we integrated hepatocellular acetaminophen metabolism, DNA damage response induction and cell fate into a multiscale mechanistic liver lobule model which involves various cell types, such as hepatocytes, residential Kupffer cells and macrophages. Our model simulations show that zonal differences in metabolism and detoxification efficiency are essential determinants of necrotic damage. Moreover, the extent of senescence, which is regulated by intracellular processes and triggered by extracellular signaling, influences the potential to recover. In silico therapies at early and late time points after APAP insult indicated that prevention of necrotic damage is most beneficial for recovery, whereas interference with regulation of senescence promotes regeneration in a less pronounced way.
Collapse
|