1
|
Wei Z, Boateng NKK, Schmitt LR, Cline H, Fernandes Fonseca MT, Newberry A, Taylor A, Adelmeijer J, Poole LG, Stravitz RT, Lee WM, Lisman T, Hansen KC, Luyendyk JP. Integrated cross-linking by TG2 and FXIII generates hepatoprotective fibrin(ogen) deposits in injured liver. Blood 2025; 145:2507-2517. [PMID: 40009455 DOI: 10.1182/blood.2024026938] [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: 09/13/2024] [Revised: 12/23/2024] [Accepted: 01/20/2025] [Indexed: 02/28/2025] Open
Abstract
ABSTRACT The transglutaminase coagulation factor XIII (FXIII) is critical for the stability and function of intravascular fibrin clots. Prorepair extravascular fibrin(ogen) deposits are potentially subject to cross-linking by FXIII and other transglutaminases not typically resident in plasma. However, the impact of these alternative modifiers on fibrin(ogen) structure and function is not known. We tested the hypothesis that tissue transglutaminase (TG2) modifies FXIII-directed fibrin(ogen) cross-linking in vitro and within injured tissue. Global proteomic analysis after experimental acetaminophen (APAP)-induced acute liver injury revealed that intrahepatic fibrin(ogen) deposition was associated with hepatic TG2 levels that exceeded that of FXIII. Mass spectrometry-based cross-link mapping of in vitro fibrin matrices uncovered, to our knowledge, the first evidence of synergistic fibrin(ogen) α-α cross-linking catalyzed by both transglutaminases. Fibrin(ogen) cross-linking was increased in livers from patients with APAP-induced acute liver failure. APAP-challenged TG2-/- mice displayed an altered pattern of FXIII-dependent fibrin(ogen)-γ and fibrin(ogen)-α chain cross-linking aligned with the impact of TG2 on fibrin cross-linking in vitro. This shift in fibrin(ogen) cross-linking exacerbated pathologies including hepatic necrosis and sinusoidal congestion. The results, to our knowledge, are the first to indicate that TG2 impacts FXIII-directed fibrin(ogen) cross-linking, both in vitro and in vivo. The results suggest that TG2 functions to dynamically alter the structure of extravascular fibrin(ogen) to mitigate liver damage, a novel mechanism likely applicable across types of tissue injury.
Collapse
Affiliation(s)
- Zimu Wei
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI
| | - Nana Kwame Kwabi Boateng
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI
| | - Lauren R Schmitt
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO
| | - Holly Cline
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI
| | | | - Ariana Newberry
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI
| | - Alicia Taylor
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI
| | - Jelle Adelmeijer
- Surgical Research Laboratory and Section of Hepatobiliary Surgery and Liver Transplantation, Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Lauren G Poole
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI
| | - R Todd Stravitz
- Section of Hepatology, Virginia Commonwealth University, Richmond, VA
| | - William M Lee
- Digestive and Liver Diseases Division, University of Texas Southwestern Medical Center, Dallas, TX
| | - Ton Lisman
- Surgical Research Laboratory and Section of Hepatobiliary Surgery and Liver Transplantation, Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO
| | - James P Luyendyk
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI
| |
Collapse
|
2
|
Wang Y, Guo J, Zhang D, Zhang X, Luo K, Gong Z. IDH1/MDH1 deacetylation activates the cGAS-STING pathway by promoting NETosis in acute liver failure. Int Immunopharmacol 2025; 158:114884. [PMID: 40388861 DOI: 10.1016/j.intimp.2025.114884] [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: 01/20/2025] [Revised: 05/12/2025] [Accepted: 05/13/2025] [Indexed: 05/21/2025]
Abstract
BACKGROUND Acute liver failure (ALF) is a life-threatening disease caused by multiple factors. Our previous study shows that IDH1/MDH1 deacetylation aggravates ALF through NETosis (a new mode of cell death). However, the exact mechanism by which NETosis mediates ALF is still unclear. METHODS The transcriptome sequencing data of hepatic tissues from ALF mice and healthy control mice were downloaded from the GEO database. Inflammation-related pathways of ALF were identified by differential expression analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. In vivo, a mouse ALF model was established with LPS/D-gal. In vitro, HepG2 cells were used to simulate hepatocyte injury. The cell death rate of HepG2 cells was detected by flow cytometry. The levels of inflammatory factors in mouse liver tissues were detected by enzyme-linked immunosorbent assay (ELISA). The formation of NETs and the levels of cGAS-STING pathway-related proteins were detected by Western blotting, immunohistochemistry and immunofluorescence. RESULTS The cGAS-STING pathway was activated in LPS/D-gal-induced ALF. In the ALF mouse model, ACY1215, as a histone deacetylase 6 inhibitor, alleviated ALF by inhibiting cGAS-STING pathway. Deoxyribonuclease 1 (DNase 1) degraded NETs formed by IDH1/MDH1 deacetylation in mouse liver tissue. DNase 1 attenuated hepatic tissue injury, reduced serum ALT and AST levels and reduced the levels of TNF-α and IL-6 in liver tissues. IDH1/MDH1 deacetylation increased the levels of cGAS-STING pathway-related protein, which was reversed by DNase 1. Exogenous NETs enhanced LPS/D-gal-induced hepatocyte death and activation of the cGAS-STING pathway in vitro. CONCLUSION IDH1/MDH1 deacetylation activates the cGAS-STING signaling pathway by NETs. The activation of STING promotes pro-inflammatory cytokine production and exacerbates the inflammatory response in LPS/D-gal-induced ALF.
Collapse
Affiliation(s)
- Yukun Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Jin Guo
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Danmei Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Xiaoya Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Ke Luo
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Zuojiong Gong
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 430060 Wuhan, China.
| |
Collapse
|
3
|
Yang B, Chen S, Xia X, Tao Z, Liu C, Li S, Zhang S, Huang J, Xia L, Quan W, Yang C, Li J. Mas Signaling Potentiates Neutropil Extracellular Traps Formation Induced by Endothelial Cells Derived S1P in Mice with Acute Liver Failure. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2411428. [PMID: 40285622 DOI: 10.1002/advs.202411428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 03/31/2025] [Indexed: 04/29/2025]
Abstract
Mas, a newly identified G-protein-coupled receptor, is prevalent in myeloid-derived immune cells and plays a key role in inflammation. This study investigates Mas signaling and neutrophil extracellular traps (NETs) in acute liver failure (ALF), aiming to elucidate their mechanisms. Male Mas1-/- and wild-type mice, aged 6-8 weeks, receive intraperitoneally injected with lipopolysaccharide (LPS)/D-galactosamine (D-Gal) (L/G) to study NETs formation. Hepatic Mas expression increases in WT-L/G mice, whereas systemic Mas1 knockout significantly reduces L/G-induced NETs and hepatotoxicity. Antibiotics treatment and co-housing (Mas1-/--L/G and WT-L/G mice) experiments show that gut flora influences the disease phenotype in Mas1-/--L/G mice. Fecal metabolite analysis suggests that mice may be protected by reduced deoxycholic acid (DCA) production in Mas1-/- activated hepatic farnesoid X receptor (FXR), suppressing sphingosine-1-phosphate (S1P)-dependent NETs. Additionally, Mas1-/- also activates the FXR-S1P-NETs axis in the liver by inhibiting SHP2. Single-cell sequencing shows decreased interaction between endothelial cells and Cldn1+CD177+ senescent neutrophils through Col4a1-CD44. This inhibits S1P-induced Raf signaling pathway activation and NETs formation. Mas signaling significantly impacts NETs formation, highlighting its potential as an anti-inflammatory therapeutic target for ALF.
Collapse
Affiliation(s)
- Bo Yang
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Shuai Chen
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Xiaoqi Xia
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Ziwen Tao
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Chun Liu
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Shanshan Li
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Shuo Zhang
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Jiali Huang
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Lu Xia
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Wenqiang Quan
- Department of Laboratory Medicine, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Changqing Yang
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Jing Li
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| |
Collapse
|
4
|
Guo J, Shi C, Wang Y, Zhang D, Zhang Q, Zhang X, Wang L, Gong Z. Targeting the HDAC6/Hint2/MICU1 axis to ameliorate acute liver failure via inhibiting NETosis. Life Sci 2025; 366-367:123498. [PMID: 39983829 DOI: 10.1016/j.lfs.2025.123498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/06/2025] [Accepted: 02/18/2025] [Indexed: 02/23/2025]
Abstract
AIMS Acute liver failure (ALF) is marked by extensive inflammation and immune dysregulation, which are closely associated with neutrophil infiltration and NETosis. However, the specific mechanisms that drive NETosis in ALF remain poorly understood. MATERIALS AND METHODS We employed flow cytometry, western blot, qRT-PCR, and cf-DNA assay to investigate the link between NETosis and ALF. The role of HDAC6-mediated deacetylation of histidine triad nucleotide-binding protein 2 (Hint2) was assessed, along with the effects of lentiviral vector-based overexpression and knockdown of Hint2 on mitochondrial function and NETosis. Additionally, CO-IP, IF, protein docking analysis, mCa2+ uptake assay, and mtROS measurement were used to explore the interaction between Hint2 and mitochondrial calcium uniporter complex (MCUc). Finally, experimental neutrophil depletion in mice was conducted to confirm the protective effect of NETosis inhibition in ALF. KEY FINDINGS Our study demonstrated that Hint2 undergoes HDAC6-mediated deacetylation, disrupting mitochondrial dynamics and triggering NETosis during ALF. Furthermore, MICU1 bridges Hint2 and NETosis by regulating mCa2+ homeostasis and mtROS production. Activation of Hint2, either through the HDAC6 inhibitor ACY1215 or via overexpression, increased the level of MICU1 to suppress the opening of the MCUc and the associated mtROS release, thereby inhibiting NETosis. Conversely, Hint2 knockdown induced NETosis by surging mCa2+ overload and mtROS production, while the MCUc inhibitor RU265 mitigates NETosis by blocking mCa2+ influx. SIGNIFICANCE Our findings recognized the HDAC6/Hint2/MICU1 axis as a novel pathway in neutrophils, the inhibition of which intercepts mCa2+ overload and mtROS accumulation, thereby reducing NETosis and facilitating liver recovery during ALF.
Collapse
Affiliation(s)
- Jin Guo
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chunxia Shi
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yukun Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Danmei Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qingqi Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoya Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Luwen Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zuojiong Gong
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
5
|
Zhu YR, Yang YQ, Ruan DD, Que YM, Gao H, Yang YZ, Zhao HJ. Paeoniflorin Attenuates APAP-Induced Liver Injury via Intervening the Crosstalk Between Hepatocyte Pyroptosis and NETs. Int J Mol Sci 2025; 26:1493. [PMID: 40003959 PMCID: PMC11855121 DOI: 10.3390/ijms26041493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/29/2025] [Accepted: 02/09/2025] [Indexed: 02/27/2025] Open
Abstract
(1) Liver injury caused by an overdose of acetaminophen (APAP) represents a major public health concern. Paeoniflorin (PF) has been reported to have anti-inflammatory and liver-protective effects, but the underlying mechanisms remain unclear. This study aimed to investigate the effect of PF on the crosstalk between pyroptosis and NETs in AILI. (2) APAP-treated C57BL/6J mice were used to demonstrate the protective effect of PF on liver injury. HepG2 and dHL-60 cells were cultured to study the effects of PF on hepatocyte pyroptosis and neutrophil extracellular traps (NETs) in vitro. Moreover, cell co-culture experiments were performed, and mice were treated with a NETs-depleting agent and hepatocyte pyroptosis inhibitor to investigate the improvement of AILI induced by PF through regulating the crosstalk between hepatocyte pyroptosis and NETs. (3) PF significantly alleviated AILI. Additionally, PF inhibited the expression of pyroptosis-related proteins, high-mobility group box 1 (HMGB1), and NETs-associated proteins in vitro and in vivo. The co-culture experiments demonstrated that PF not only inhibited the NETs triggered by hepatocyte pyroptosis, but also suppressed the hepatocyte pyroptosis induced by NETs. In mice with depleted neutrophils, the level of hepatocyte pyroptosis notably decreased, indicating a diminished impact of PF. Similarly, NETs formation was reduced in mice receiving a pyroptosis inhibitor compared to the APAP group. Compared with DNase I alone, the reduction effect of PF combined with DNase I on serum ALT and AST levels decreased from 46.857% and 39.927% to 44.347% and 33.419%, respectively. Compared with DSF alone, PF combined with DSF reduced the ALT and AST levels from 46.857% and 39.927% to 45.347% and 36.419%, respectively. (4) PF demonstrated therapeutic effects on AILI. Its mechanism involves the regulation of the crosstalk between hepatocyte pyroptosis and NETs. This research substantiates the pharmacological promise of PF as a therapeutic intervention for acute AILI.
Collapse
Affiliation(s)
- Yu-Ru Zhu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311402, China; (Y.-R.Z.); (Y.-Q.Y.); (D.-D.R.); (Y.-M.Q.)
| | - Ya-Qin Yang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311402, China; (Y.-R.Z.); (Y.-Q.Y.); (D.-D.R.); (Y.-M.Q.)
| | - Dan-Dan Ruan
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311402, China; (Y.-R.Z.); (Y.-Q.Y.); (D.-D.R.); (Y.-M.Q.)
| | - Yue-Mei Que
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311402, China; (Y.-R.Z.); (Y.-Q.Y.); (D.-D.R.); (Y.-M.Q.)
| | - Hang Gao
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China;
| | - Yan-Zi Yang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311402, China; (Y.-R.Z.); (Y.-Q.Y.); (D.-D.R.); (Y.-M.Q.)
| | - Hua-Jun Zhao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311402, China; (Y.-R.Z.); (Y.-Q.Y.); (D.-D.R.); (Y.-M.Q.)
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China;
| |
Collapse
|
6
|
McGettigan B, Hernandez-Tejero M, Malhi H, Shah V. Immune Dysfunction and Infection Risk in Advanced Liver Disease. Gastroenterology 2025:S0016-5085(24)05694-4. [PMID: 39927926 DOI: 10.1053/j.gastro.2024.08.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 02/11/2025]
Abstract
The risk of microbial infections is increased in cirrhosis and other forms of advanced liver disease such as alcohol-associated hepatitis. Such infections may precipitate new or further decompensation and death, especially in patients with clinical features of acute-on-chronic liver failure. The severe immune dysfunction or "immune paralysis" caused by advanced liver disease is associated with high short-term mortality. However, the pathogenic mechanisms underlying immune dysfunction and immunodeficiency are incompletely understood. Evidence to date suggests a complex, dynamic process that perturbs the physiological roles of the liver as a master regulator of systemic immunity and protector against noxious effects of exogenous molecules in the portal vein flowing from the gut. Thus, in cirrhosis and severe alcohol-associated hepatitis, the ability of hepatocytes and intrahepatic immune cells to balance normal context-dependent dichotomous responses of tolerance vs immune activation is lost. Contributing factors include loss of the gut barrier with translocation of microbial products through the portal vein, culminating in development of functional defects in innate and adaptive immune cells, and generation of immune-regulatory myeloid cells that permit microbial colonization and infection. This review addresses key evidence supporting the paradigm of immune dysfunction as a risk for microbial infections and identifies potential therapeutic targets for intervention. The primary focus is on cirrhosis-associated immune dysfunction and alcohol-associated liver disease, because the bulk of available data are from these 2 conditions.
Collapse
Affiliation(s)
- Brett McGettigan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Maria Hernandez-Tejero
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Harmeet Malhi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Vijay Shah
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
7
|
Zuo J, Tian YX, An Q, Wu BY, Yang JR, Fan YC. Potential Biomarkers and Therapeutic Targets in Hepatitis B Virus-related Acute Liver Failure: Interplay of the Ferroptosis, Autophagy and Immune Responses. Int J Med Sci 2025; 22:806-818. [PMID: 39991755 PMCID: PMC11843133 DOI: 10.7150/ijms.106360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/11/2025] [Indexed: 02/12/2025] Open
Abstract
Hepatitis B virus-related acute liver failure (HBV-ALF) is characterized by a high fatality rate, its pathogenesis remains unclear and the therapeutic efficacy is limited. Ferroptosis which closely related to autophagy may be an underlying mechanism of HBV-ALF. The aim of this study was to identify key ferroptosis- and autophagy-related genes and pathways and provide insight into potential therapeutic approaches for HBV-ALF. We accessed the GSE14668 and GSE96851 datasets from the Gene Expression Omnibus (GEO) database and focused on differentially expressed genes (DEGs), ferroptosis-related DEGs (FRGs) and autophagy-related DEGs (ARGs). Hub genes were subsequently analyzed for enrichment, protein‒protein interactions (PPIs), and different immunological microenvironments, and potential hub gene were identified using MCC method and LASSO. Gene-targeted drugs were from the DGIdb and DrugBank databases.A total of 1462 DEGs were identified (726 upregulated and 736 downregulated). Enriched pathways included amino acid metabolism and immune and inflammatory responses, potentially serving as biomarkers for ALF pathogenesis. After integration with the FerrDb and HADb databases, 55 FRGs and 45 ARGs were identified. Thirteen hub genes (SLC7A11, HMOX1, G6PD, RRM2, KIF20A, HELLS, GPT2, GLS2, SPP1, CCR2, DCN, IRS1, and IGF1) were identified which closely associated with the immune microenvironment. Interplay among these genes occurred primarily through HMOX1. Moreover, we identified several hub gene-targeted drugs that may be effective in HBV-ALF treatment, such as riluzole, acetylcysteine, NADH and Vitamin E.Thirteen hub genes may play crucial roles in HBV-ALF progression, particularly, the HMOX1. Furthermore, drug target exploration offered promising avenues for therapeutic intervention in patients with HBV-ALF.
Collapse
Affiliation(s)
- Jing Zuo
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
- Institute of Hepatology, Shandong University, Jinan, China
| | - Yu-Xin Tian
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
- Institute of Hepatology, Shandong University, Jinan, China
| | - Qi An
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
- Institute of Hepatology, Shandong University, Jinan, China
| | - Bai-Yun Wu
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
- Institute of Hepatology, Shandong University, Jinan, China
| | - Jie-Ru Yang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
- Institute of Hepatology, Shandong University, Jinan, China
| | - Yu-Chen Fan
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
- Institute of Hepatology, Shandong University, Jinan, China
| |
Collapse
|
8
|
von Meijenfeldt FA, Lisman T, Pacheco A, Zen Y, Bernal W. Histologic evidence of neutrophil extracellular traps and fibrin(ogen) deposition in liver biopsies from patients with inflammatory liver disease. Res Pract Thromb Haemost 2025; 9:102666. [PMID: 39959636 PMCID: PMC11830338 DOI: 10.1016/j.rpth.2024.102666] [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: 09/16/2024] [Revised: 12/12/2024] [Accepted: 12/20/2024] [Indexed: 02/18/2025] Open
Abstract
Background Liver disease is often characterized by the activation of coagulation and inflammation. Experimental studies suggest that the interaction between neutrophils and platelets with local activation of coagulation could contribute to liver injury progression, but there have been limited studies in humans. Objectives We studied the hemostatic components and neutrophil extracellular traps (NETs) in liver biopsies from patients with different inflammatory liver diseases. Methods Liver biopsies from patients with inflammatory liver disease (alcoholic steatohepatitis [ASH], autoimmune hepatitis, primary sclerosing cholangitis, metabolic-associated steatohepatitis, and allograft ischemia-reperfusion injury (IRI), each n = 20) were stained for fibrin(ogen), platelets, and NETs. The correlation of NET formation with deposition of hemostatic components and laboratory measures of disease severity was investigated. Results In 75% of the liver biopsies, no fibrin(ogen) was detectable, and only 20% of the biopsies showed minimal deposition. Overall, 50% of liver biopsies stained positive for NETs. Platelet deposition and NET formation were highest in IRI, where it correlated with histologic severity of injury (r = .61 [95% CI, .22-.84]; P < .01) and ASH. Platelet deposition was associated with NET formation (r = .44 [95% CI, .27-.59]; P < .001) and colocalized in the biopsies. NET formation, but not fibrin and platelet deposition, was moderately associated with the model of end-stage liver disease score (r = .29 [95% CI, .07-.49]; P < .01). Conclusion In contrast to experimental studies, we demonstrated minimal intrahepatic fibrin(ogen) deposition in different types of human inflammatory liver disease. Histologic evidence for intrahepatic NETs was common and most pronounced in acute ASH and IRI and was associated with platelet deposition and disease severity.
Collapse
Affiliation(s)
- Fien A. von Meijenfeldt
- Surgical Research Laboratory, Department of Surgery, University Medical Center Groningen, Groningen, the Netherlands
| | - Ton Lisman
- Surgical Research Laboratory, Department of Surgery, University Medical Center Groningen, Groningen, the Netherlands
| | - Alessandra Pacheco
- Institute of Liver Studies, King’s College Hospital, London, United Kingdom
| | - Yoh Zen
- Institute of Liver Studies, King’s College Hospital, London, United Kingdom
| | - William Bernal
- Institute of Liver Studies, King’s College Hospital, London, United Kingdom
- Liver Intensive Care Unit, Institute of Liver Studies, King’s College Hospital, London, United Kingdom
| |
Collapse
|
9
|
Wang Y, Guo J, Zhang D, Shi C, Zhang X, Gong Z. IDH1/MDH1 deacetylation promotes NETosis by regulating OPA1 and autophagy. Int Immunopharmacol 2024; 143:113270. [PMID: 39353390 DOI: 10.1016/j.intimp.2024.113270] [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: 08/15/2024] [Revised: 09/21/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND As a heterogeneous and life-threatening disease, the pathogenesis of acute liver failure (ALF) is complex. Our previous study has shown that IDH1/MDH1 deacetylation promotes ALF by regulating NETosis (a novel mode of cell death). In this article, we explore the manners of IDH1/MDH1 deacetylation regulates NETosis. METHODS In vitro experiments, the formation of NETs was detected by immunofluorescence staining and Western blotting. LC3 fluorescence staining was used to detect autophagosome formation. To observe mitochondrial morphology, cells were stained by Mito-Tracker Red. Western blotting was used to detect the levels of autophagy protein and mitochondrial dynamin. In vivo experiments, the ALF model in mouse was established with LPS/D-gal, and the formation of NETs was detected by immunofluorescence staining and Western blotting. The autophagy levels were detected by Western blotting in liver samples. RESULTS In dHL-60 cells, Western blotting results showed that the expression of OPA1 was higher in the IDH1/MDH1 deacetylated group compared with the IDH1/MDH1 WT group. And histone deacetylase inhibitor 6 (HDAC6i, ACY1215) decreased the expression level of OPA1 in IDH1/MDH1 deacetylated group. IDH1/MDH1 deacetylation increased the expression levels of both LC3B-II and Beclin 1, while decreasing the expression level of P62. It was reversed by ACY1215. Combined with our previous experiments, IDH1/MDH1 deacetylation upregulated autophagy concomitant with the increased expression of the markers of NETs formation. In a mouse model of ALF, ACY1215 further decreased the expression levels of LC3B-II and Beclin 1, while increasing the expression level of P62 in IDH1/MDH1 deacetylated mice. CONCLUSIONS IDH1/MDH1 deacetylation promoted NETosis by regulating autophagy and OPA1 in vitro. The regulation of neutrophil autophagy on NETosis during IDH1/MDH1 deacetylation might be masked in mice. ACY1215 might attenuate NETosis by regulating neutrophil autophagy, which alleviated ALF aggravated by IDH1/MDH1 deacetylation.
Collapse
Affiliation(s)
- Yukun Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Jin Guo
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Danmei Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Chunxia Shi
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Xiaoya Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Zuojiong Gong
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 430060 Wuhan, China.
| |
Collapse
|
10
|
Ye R, Wei Y, Li J, Xu M, Xie H, Huang J, Deng L, Li C. MiRNAs and Neutrophil-Related Membrane Proteins from Plasma-Derived Extracellular Vesicles for Early Prediction of Organ Dysfunction and Prognosis in Septic Patients. J Inflamm Res 2024; 17:10347-10369. [PMID: 39649421 PMCID: PMC11625425 DOI: 10.2147/jir.s492902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 11/28/2024] [Indexed: 12/10/2024] Open
Abstract
Purpose The pathogenesis of sepsis-induced organ dysfunction remains elusive, and the mortality remains alarmingly high. We sought to investigate the profile of extracellular vesicles (EVs)-mediated communication between plasma and polymorphonuclear neutrophils (PMNs) in sepsis, and to elucidate whether miRNAs and PMN-related membrane proteins from plasma-derived EVs (plasma-EVs) are associated with sepsis-induced organ dysfunction and prognosis. Methods PMN-derived EVs (PMN-EVs) were isolated from the blood samples of healthy controls (N=3) and patients with septic shock (N=3) after ICU admission. We performed miRNA sequencing of the isolated EVs, followed by bioinformatic analysis. A miRNA model for comparing PMN-EVs and plasma-EVs was successfully established in the training cohort. Furthermore, miRNAs and PMN-related membrane proteins from the plasma-EV model were confirmed in the validation cohort. A logistic regression model, receiver operating characteristic (ROC) curves, and Kaplan-Meier analyses were performed to evaluate the efficiency of diagnostic and/or prognostic performance. Further, in vivo and in vitro experiments were conducted to explore the involvement of plasma-EVs in PMNs autophagy. Results Fifty-five miRNAs from PMN-EVs differed significantly between the healthy controls and patients with septic shock. Furthermore, the plasma-EV model (six miRNAs and eight PMN-related membrane proteins) was confirmed in the validation cohort, demonstrating that miR-34a-5p, miR-503-5p, miR-4772-3p, ITGAM, MPO, and MMP9 serve as sepsis biomarkers for distinguishing lung, liver, and kidney dysfunction. Kaplan-Meier survival analysis showed that miR-34a-5p, miR-4772-3p, ITGAM, and MMP9 were potential prognostic predictors. Finally, we found that plasma-EVs from sepsis patients exert an inhibitory effect on PMNs autophagy, which can be reversed by EV inhibitors such as GW4869 and enoxaparin. Conclusion These findings suggest that miRNAs and PMN-related membrane proteins from plasma-EVs could be valuable diagnostic tools for identifying sepsis-induced organ dysfunction and predicting prognosis, enabling proactive management of sepsis by physicians and improving the prognosis of sepsis patients.
Collapse
Affiliation(s)
- Rongzong Ye
- Department of Emergency Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Yating Wei
- Department of Emergency Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Jingwen Li
- Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Meili Xu
- Department of Emergency Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Haiyang Xie
- Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Jiahao Huang
- Department of Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524000, People’s Republic of China
| | - Liehua Deng
- Department of Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524000, People’s Republic of China
| | - Chaoqian Li
- Department of Emergency Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| |
Collapse
|
11
|
Mittendorfer M, Pierre L, Huzevka T, Schofield J, Abrams ST, Wang G, Toh CH, Bèchet NB, Caprnja I, Kjellberg G, Aswani A, Olm F, Lindstedt S. Restoring discarded porcine lungs by ex vivo removal of neutrophil extracellular traps. J Heart Lung Transplant 2024; 43:1919-1929. [PMID: 39038563 DOI: 10.1016/j.healun.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 06/16/2024] [Accepted: 07/08/2024] [Indexed: 07/24/2024] Open
Abstract
BACKGROUND By causing inflammation and tissue damage, neutrophil extracellular traps (NETs) constitute an underlying mechanism of aspiration-induced lung injury, a major factor of the low utilization of donor lungs in lung transplantation (LTx). METHODS To determine whether NET removal during ex vivo lung perfusion (EVLP) can restore lung function and morphology in aspiration-damaged lungs, gastric aspiration lung injury was induced in 12 pigs. After confirmation of acute respiratory distress syndrome, the lungs were explanted and assigned to NET removal connected to EVLP (treated) (n = 6) or EVLP only (nontreated) (n = 6). Hemodynamic measurements were taken, and blood and tissue samples were collected to assess lung function, morphology, levels of cell-free DNA, extracellular histones, and nucleosomes as markers of NETs, as well as cytokine levels. RESULTS After EVLP and NET removal in porcine lungs, PaO2/FiO2 ratios increased significantly compared to those undergoing EVLP alone (p = 0.0411). Treated lungs had lower cell-free DNA (p = 0.0260) and lower levels of extracellular histones in EVLP perfusate (p= 0.0260) than nontreated lungs. According to histopathology, treated lungs showed less immune cell infiltration and less edema compared with nontreated lungs, which was reflected in decreased levels of proinflammatory cytokines in EVLP perfusate and bronchoalveolar lavage fluid. CONCLUSIONS To conclude, removing NETs during EVLP improved lung function and morphology in aspiration-damaged donor lungs. The ability to remove NETs during EVLP could represent a new therapeutic approach for LTx and potentially expand the donor pool for transplantation.
Collapse
Affiliation(s)
- Margareta Mittendorfer
- Department of Clinical Sciences, Lund University, Lund, Sweden; Department of Cardiothoracic Surgery and Transplantation, Lund University Hospital, Lund, Sweden; Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden
| | - Leif Pierre
- Department of Cardiothoracic Surgery and Transplantation, Lund University Hospital, Lund, Sweden; Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden
| | - Tibor Huzevka
- Department of Clinical Sciences, Lund University, Lund, Sweden; Department of Cardiothoracic Surgery and Transplantation, Lund University Hospital, Lund, Sweden; Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden
| | - Jeremy Schofield
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Simon T Abrams
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Guozheng Wang
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Cheng-Hock Toh
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom; Roald Dahl Haemostasis & Thrombosis Centre, Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| | - Nicholas B Bèchet
- Department of Clinical Sciences, Lund University, Lund, Sweden; Department of Cardiothoracic Surgery and Transplantation, Lund University Hospital, Lund, Sweden; Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden
| | - Ilma Caprnja
- Department of Clinical Sciences, Lund University, Lund, Sweden; Department of Cardiothoracic Surgery and Transplantation, Lund University Hospital, Lund, Sweden; Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden
| | - Gunilla Kjellberg
- Department of Thoracic Surgery and Anaesthesiology, Uppsala University Hospital, Uppsala, Sweden
| | - Andrew Aswani
- Department of Critical Care, Guy's and St Thomas's NHS Foundation Trust, London, United Kingdom; Santersus AG, Zurich, Switzerland
| | - Franziska Olm
- Department of Clinical Sciences, Lund University, Lund, Sweden; Department of Cardiothoracic Surgery and Transplantation, Lund University Hospital, Lund, Sweden; Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden
| | - Sandra Lindstedt
- Department of Clinical Sciences, Lund University, Lund, Sweden; Department of Cardiothoracic Surgery and Transplantation, Lund University Hospital, Lund, Sweden; Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden.
| |
Collapse
|
12
|
Qiu S, Zhao Y, Hu J, Zhang Q, Wang L, Chen R, Cao Y, Liu F, Zhao C, Zhang L, Ren W, Xin S, Chen Y, Duan Z, Han T. Predicting the 28-day prognosis of acute-on-chronic liver failure patients based on machine learning. Dig Liver Dis 2024; 56:2095-2102. [PMID: 39004553 DOI: 10.1016/j.dld.2024.06.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/22/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND We aimed to establish a prognostic predictive model based on machine learning (ML) methods to predict the 28-day mortality of acute-on-chronic liver failure (ACLF) patients, and to evaluate treatment effectiveness. METHODS ACLF patients from six tertiary hospitals were included for analysis. Features for ML models' development were selected by LASSO regression. Models' performance was evaluated by area under the curve (AUC) and accuracy. Shapley additive explanation was used to explain the ML model. RESULTS Of the 736 included patients, 587 were assigned to a training set and 149 to an external validation set. Features selected included age, hepatic encephalopathy, total bilirubin, PTA, and creatinine. The eXtreme Gradient Boosting (XGB) model outperformed other ML models in the prognostic prediction of ACLF patients, with the highest AUC and accuracy. Delong's test demonstrated that the XGB model outperformed Child-Pugh score, MELD score, CLIF-SOFA, CLIF-C OF, and CLIF-C ACLF. Sequential assessments at baseline, day 3, day 7, and day 14 improved the predictive performance of the XGB-ML model and can help clinicians evaluate the effectiveness of medical treatment. CONCLUSIONS We established an XGB-ML model to predict the 28-day mortality of ACLF patients as well as to evaluate the treatment effectiveness.
Collapse
Affiliation(s)
- Shaotian Qiu
- The School of Medicine, Nankai University, Tianjin 300071, China; Department of Gastroenterology and Hepatology, Tianjin Union Medical Center Affiliated to Nankai University, Tianjin 300121, China
| | - Yumeng Zhao
- The School of Medicine, Nankai University, Tianjin 300071, China
| | - Jiaxuan Hu
- The School of Medicine, Nankai University, Tianjin 300071, China; Department of Gastroenterology and Hepatology, Tianjin Union Medical Center Affiliated to Nankai University, Tianjin 300121, China
| | - Qian Zhang
- Department of Gastroenterology and Hepatology, Tianjin Union Medical Center, Tianjin 300121, China; Tianjin Medical University, Tianjin 300070, China
| | - Lewei Wang
- Department of Gastroenterology and Hepatology, Tianjin Union Medical Center of Tianjin Medical University, Tianjin 300121, China
| | - Rui Chen
- Department of Gastroenterology and Hepatology, Tianjin Union Medical Center of Tianjin Medical University, Tianjin 300121, China
| | - Yingying Cao
- Department of Hepatology and Gastroenterology, The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China
| | - Fang Liu
- Department of Hepatology and Gastroenterology, The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China
| | - Caiyan Zhao
- Department of Infectious Disease, the Third Hospital of Hebei Medical University, Shijiazhuang 050051, China
| | - Liaoyun Zhang
- Department of Infection Disease, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Wanhua Ren
- Infectious Department of Shandong First Medical University Affiliated Shandong Provincial Hospital, Jinan 250021, China
| | - Shaojie Xin
- Liver Failure Treatment and Research Center, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Yu Chen
- Liver Disease Center (Difficult & Complicated Liver Diseases and Artificial Liver Center), Beijing You'an Hospital Affiliated to Capital Medical University, Beijing 100069, China
| | - Zhongping Duan
- Liver Disease Center (Difficult & Complicated Liver Diseases and Artificial Liver Center), Beijing You'an Hospital Affiliated to Capital Medical University, Beijing 100069, China
| | - Tao Han
- The School of Medicine, Nankai University, Tianjin 300071, China; Department of Gastroenterology and Hepatology, Tianjin Union Medical Center Affiliated to Nankai University, Tianjin 300121, China; Department of Gastroenterology and Hepatology, Tianjin Union Medical Center, Tianjin 300121, China; Tianjin Medical University, Tianjin 300070, China; Department of Gastroenterology and Hepatology, Tianjin Union Medical Center of Tianjin Medical University, Tianjin 300121, China; Department of Hepatology and Gastroenterology, The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China.
| |
Collapse
|
13
|
Zhang Y, Luo Q, Lin X, Wang L, Li Z, Chen J, Xu R, Wu L, Peng L, Xu W. Development and Validation of a New Model Including Inflammation Indexes for the Long-Term Prognosis of Hepatitis B-Related Acute-On-Chronic Liver Failure. J Med Virol 2024; 96:e70110. [PMID: 39651596 DOI: 10.1002/jmv.70110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/30/2024] [Accepted: 11/23/2024] [Indexed: 12/11/2024]
Abstract
Acute-on-chronic liver failure (ACLF) is a severe condition characterized by a systemic inflammatory response and associated with high mortality. Currently, there is no reliable prediction model for long-term prognosis in ACLF. This study aimed to develop and validate a prognostic model incorporating inflammation indexes to predict the long-term outcome of patients with hepatitis B virus-related ACLF (HBV-ACLF). A retrospective analysis of clinical data from HBV-ACLF patients (n = 986) treated at the Third Affiliated Hospital of Sun Yat-sen University between January 2014 and December 2018 was conducted. Patients were randomly divided into training (n = 690) and validation (n = 296) cohorts. The Least Absolute Shrinkage and Selection Operator (LASSO) and Cox regression analyses were used to identify independent risk factors for long-term mortality. The following variables were identified as independent predictors of long-term mortality: age, cirrhosis, hepatic encephalopathy, total bilirubin (TBIL), international normalized ratio (INR), monocyte-to-lymphocyte ratio (MLR), and neutrophil-to-platelet ratio (NPR). A novel nomogram was established by assigning weights to each variable. The C-index of the nomogram was 0.777 (95% confidence interval [CI]: 0.752-0.802). In the training set, the area under the curve (AUC) for predicting mortality at 1, 3, and 12 months was 0.841 (95% CI: 0.807-0.875), 0.827 (95% CI: 0.796-0.859), and 0.829 (95% CI: 0.798-0.859), respectively. The nomogram demonstrated superior predictive performance for 12-month survival compared to the model for end-stage liver disease (MELD) score (0.767, 95% CI: 0.730-0.804, p < 0.001) and the clinical overt sepsis in acute liver failure clinical practice Guidelines-ACLF II score (0.807, 95% CI: 0.774-0.840, p = 0.028). Finally, calibration curves and decision curve analysis (DCA) confirmed the clinical utility of the nomogram. The novel inflammation-based scoring system, incorporating MLR and NPR, effectively predicts long-term mortality in HBV-ACLF patients.
Collapse
Grants
- This study was supported by grants from the Natural Science Foundation of China (No. 82070611 to Liang Peng), Natural Science Foundation of Guangdong Province (No. 2020A1515010317 to Liang Peng), GuangDong Basic and Applied Basic Research Foundation (No. 21202104030000608 and 2021A1515220029 to Liang Peng), Guangzhou Science and Technology Plan Projects (No. 202102010204 and 2023B03J1287 to Liang Peng, and No. 202102080064 to Wenxiong Xu), Sun Yat-Sen University Clinical Research 5010 Program (No. 2020007 and 2018009 to Liang Peng), the Five-Year Plan of Third Affiliated Hospital of Sun Yat-sen University (No. K00006 and P02421 to Liang Peng), and Beijing iGandan Foundation (No. iGandanF-1082022-RGG038 to Wenxiong Xu and No. iGandanF-1082024-RGG050 to Liang Peng). All funders did not participate in the design of the study, collection, analysis, and interpretation of data, as well as in writing the manuscript.
Collapse
Affiliation(s)
- Yeqiong Zhang
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qiumin Luo
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiumei Lin
- Department of Clinical Laboratory, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lu Wang
- Department of Diagnostics, Second School of Clinical Medicine, Binzhou Medical University, Yantai, China
| | - Zhipeng Li
- Department of Emergency, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jia Chen
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ruixuan Xu
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lina Wu
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Liang Peng
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wenxiong Xu
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
14
|
Li S, Xu G, Guo Z, Liu Y, Ouyang Z, Li Y, Huang Y, Sun Q, Giri BR, Fu Q. Deficiency of hasB accelerated the clearance of Streptococcus equi subsp. Zooepidemicus through gasdermin d-dependent neutrophil extracellular traps. Int Immunopharmacol 2024; 140:112829. [PMID: 39083933 DOI: 10.1016/j.intimp.2024.112829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024]
Abstract
Streptococcus equi subsp. zooepidemicus (S. zooepidemicus, SEZ) is an essential zoonotic bacterial pathogen that can cause various inflammation, such as meningitis, endocarditis, and pneumonia. UDP-glucose dehydrogenase (hasB) is indispensable in synthesizing SEZ virulence factor hyaluronan capsules. Our study investigated the infection of hasB on mice response to SEZ by employing a constructed capsule-deficient mutant strain designated as the ΔhasB strain. This deficiency was associated with a reduced SEZ bacterial load in the mice's blood and peritoneal lavage fluid (PLF) post-infection. Besides, the ΔhasB SEZ strain exhibited a higher propensity for neutrophil infiltration and release of cell-free DNA (cfDNA) in vivo compared to the wild-type (WT) SEZ strain. In vitro experiments further revealed that ΔhasB SEZ more effectively induced the formation of neutrophil extracellular traps (NETs) containing histone 3 (H3), neutrophil elastase (NE), and DNA, than its WT counterpart. Moreover, the release of NETs was determined to be gasdermin D (GSDMD)-dependent during the infection process. Taken together, these findings underscore that the deficiency of the hasB gene in SEZ leads to enhanced GSDMD-dependent NET release from neutrophils, thereby reducing SEZ's capacity to resist NETs-mediated eradication during infection. Our finding paves the way for the development of innovative therapeutic strategies against SEZ.
Collapse
Affiliation(s)
- Shun Li
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China; Foshan University Veterinary Teaching Hospital, Foshan University, Foshan, Guangdong, China
| | - Guobin Xu
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China
| | - Zheng Guo
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China
| | - Yuxuan Liu
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China
| | - Zhiliang Ouyang
- Houjie Town Agricultural Technology Service Center, Dongguan, Guangdong, China
| | - Yajuan Li
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China; Foshan University Veterinary Teaching Hospital, Foshan University, Foshan, Guangdong, China
| | - Yunfei Huang
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China; Foshan University Veterinary Teaching Hospital, Foshan University, Foshan, Guangdong, China
| | - Qinqin Sun
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China; Foshan University Veterinary Teaching Hospital, Foshan University, Foshan, Guangdong, China
| | - Bikash R Giri
- Department of Zoology, K.K.S. Women's College, Balasore, Odisha, India
| | - Qiang Fu
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China; Foshan University Veterinary Teaching Hospital, Foshan University, Foshan, Guangdong, China.
| |
Collapse
|
15
|
Zhang K, Jia R, Zhang Q, Xiang S, Wang N, Xu L. Metabolic dysregulation-triggered neutrophil extracellular traps exacerbate acute liver failure. FEBS Lett 2024; 598:2450-2462. [PMID: 39155145 DOI: 10.1002/1873-3468.14971] [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: 04/24/2024] [Accepted: 05/27/2024] [Indexed: 08/20/2024]
Abstract
Acute liver failure (ALF) is an acute liver disease with a high mortality rate in clinical practice, characterized histologically by extensive hepatocellular necrosis and massive neutrophil infiltration. However, the role of these abnormally infiltrating neutrophils during ALF development is unclear. Here, in an ALF mouse model, metabolites were identified that promote the formation of neutrophil extracellular traps (NETs) in the liver, subsequently influencing macrophage differentiation and disease progression. ALF occurs with abnormalities in hepatic and intestinal metabolites. Abnormal metabolites (LTD4 and glutathione) can directly, or indirectly via reactive oxygen species, promote NET formation of infiltrating neutrophils, which subsequently regulate macrophages in a pro-inflammatory M1-like state, inducing an amplification of the destructive effects of inflammation. Together, this study provides new insights into the role of NETs in the pathogenesis of ALF.
Collapse
Affiliation(s)
- Kangnan Zhang
- Department of Gastroenterology, Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, China
| | - Rongrong Jia
- Department of Gastroenterology, Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, China
| | - Qinghui Zhang
- Department of Clinical Laboratory, Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, China
| | - Shihao Xiang
- Department of Gastroenterology, Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, China
| | - Na Wang
- Department of Gastroenterology, Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, China
| | - Ling Xu
- Department of Gastroenterology, Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, China
| |
Collapse
|
16
|
Zhang D, Guo J, Shi C, Wang Y, Zhang Y, Zhang X, Gong Z. MPO-DNA Complexes and cf-DNA in Patients with Sepsis and Their Clinical Value. Biomedicines 2024; 12:2190. [PMID: 39457503 PMCID: PMC11505433 DOI: 10.3390/biomedicines12102190] [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: 09/09/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Neutrophils, as the first line of defense in the immune response, produce neutrophil extracellular traps (NETs) upon activation, which are significant in the pathogenesis and organ damage in sepsis. This study aims to explore the clinical value of myeloperoxidase-DNA (MPO-DNA) and cell-free DNA (cf-DNA) in sepsis patients. Methods: Clinical data were collected from 106 sepsis patients, 25 non-sepsis patients, and 51 healthy controls. Sequential Organ Failure Assessment (SOFA) scores were calculated, and levels of MPO-DNA) complexes and cf-DNA were measured using specific kits. Correlation analyses assessed relationships between indicators, while logistic regression identified independent risk factors. Receiver operating characteristic (ROC) curves calculated the area under the curve (AUC) to evaluate the diagnostic value of the biomarkers. Results: Sepsis patients exhibited significantly elevated levels of MPO-DNA and cf-DNA compared to non-sepsis patients and healthy controls. In sepsis patients, MPO-DNA and cf-DNA levels correlated with inflammation, coagulation, and organ damage indicators, as well as procalcitonin (PCT) levels and SOFA scores. Both C-reactive protein (CRP) and cf-DNA were identified as independent risk factors for sepsis, demonstrating moderate diagnostic value. ROC analysis showed that the combination of MPO-DNA and CRP (AUC: 0.837) enhances the AUC value of CRP (0.777). Conclusions: In summary, elevated serum levels of MPO-DNA and cf-DNA in sepsis patients correlate with SOFA scores and PCT levels, providing reference value for sepsis diagnosis in clinical settings.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Zuojiong Gong
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan 430060, China; (D.Z.); (J.G.); (Y.W.); (Y.Z.); (X.Z.)
| |
Collapse
|
17
|
Wang H, Kim SJ, Lei Y, Wang S, Wang H, Huang H, Zhang H, Tsung A. Neutrophil extracellular traps in homeostasis and disease. Signal Transduct Target Ther 2024; 9:235. [PMID: 39300084 PMCID: PMC11415080 DOI: 10.1038/s41392-024-01933-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 06/25/2024] [Accepted: 07/16/2024] [Indexed: 09/22/2024] Open
Abstract
Neutrophil extracellular traps (NETs), crucial in immune defense mechanisms, are renowned for their propensity to expel decondensed chromatin embedded with inflammatory proteins. Our comprehension of NETs in pathogen clearance, immune regulation and disease pathogenesis, has grown significantly in recent years. NETs are not only pivotal in the context of infections but also exhibit significant involvement in sterile inflammation. Evidence suggests that excessive accumulation of NETs can result in vessel occlusion, tissue damage, and prolonged inflammatory responses, thereby contributing to the progression and exacerbation of various pathological states. Nevertheless, NETs exhibit dual functionalities in certain pathological contexts. While NETs may act as autoantigens, aggregated NET complexes can function as inflammatory mediators by degrading proinflammatory cytokines and chemokines. The delineation of molecules and signaling pathways governing NET formation aids in refining our appreciation of NETs' role in immune homeostasis, inflammation, autoimmune diseases, metabolic dysregulation, and cancer. In this comprehensive review, we delve into the multifaceted roles of NETs in both homeostasis and disease, whilst discussing their potential as therapeutic targets. Our aim is to enhance the understanding of the intricate functions of NETs across the spectrum from physiology to pathology.
Collapse
Affiliation(s)
- Han Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Susan J Kim
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Yu Lei
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuhui Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hui Wang
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hai Huang
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Hongji Zhang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| | - Allan Tsung
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
18
|
Zhang Y, Shi K, Zhu B, Feng Y, Liu Y, Wang X. Neutrophil Extracellular Trap Scores Predict 90-Day Mortality in Hepatitis B-Related Acute-on-Chronic Liver Failure. Biomedicines 2024; 12:2048. [PMID: 39335563 PMCID: PMC11429194 DOI: 10.3390/biomedicines12092048] [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: 08/14/2024] [Revised: 08/30/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Hepatitis B virus (HBV)-related acute-on-chronic liver failure (ACLF) is associated with pronounced systemic inflammation, and neutrophil extracellular traps (NETs) are key components of this response. The primary objective of this study was to establish an NET-related scoring system for patients with HBV-ACLF. A prospective training cohort of 81 patients from the Beijing Ditan Hospital was included. The concentrations of NET markers (cell-free DNA, myeloperoxidase DNA [MPO-DNA], and citrullinated histone H3) in peripheral blood were quantified. Random survival forest, LASSO regression, and multivariate Cox regression analyses were used to identify prognostic factors associated with 90-day mortality in ACLF patients and develop a nomogram for visualization, which was followed by evaluation in a validation cohort (n = 40). NET-related marker levels were significantly higher in the non-survival group than in the survival group (p < 0.05). The NET score was constructed by combining MPO-DNA, neutrophil-to-lymphocyte ratio, and age data. The score's diagnostic effectiveness, assessed by the area under the curve, yielded values of 0.83 and 0.77 in the training and validation sets, respectively, markedly surpassing those of other established models (p < 0.05). In both groups, the 90-day mortality rates were 88.8% and 75.0%, respectively, for patients categorized as high risk and 18.0% and 12.5%, respectively, for those classified as low risk.
Collapse
Affiliation(s)
| | | | | | | | | | - Xianbo Wang
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing 100054, China; (Y.Z.); (K.S.); (B.Z.); (Y.F.); (Y.L.)
| |
Collapse
|
19
|
Sun J, Yang F, Zheng Y, Huang C, Fan X, Yang L. Pathogenesis and interaction of neutrophils and extracellular vesicles in noncancer liver diseases. Int Immunopharmacol 2024; 137:112442. [PMID: 38889508 DOI: 10.1016/j.intimp.2024.112442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/20/2024]
Abstract
Liver disease ranks as the eleventh leading cause of mortality, leading to approximately 2 million deaths annually worldwide. Neutrophils are a type of immune cell that are abundant in peripheral blood and play a vital role in innate immunity by quickly reaching the site of liver injury. They exert their influence on liver diseases through autocrine, paracrine, and immunomodulatory mechanisms. Extracellular vesicles, phospholipid bilayer vesicles, transport a variety of substances, such as proteins, nucleic acids, lipids, and pathogenic factors, for intercellular communication. They regulate cell communication and perform their functions by delivering biological information. Current research has revealed the involvement of the interaction between neutrophils and extracellular vesicles in the pathogenesis of liver disease. Moreover, more research has focused on targeting neutrophils as a therapeutic strategy to attenuate disease progression. Therefore, this article summarizes the roles of neutrophils, extracellular vesicles, and their interactions in noncancerous liver diseases.
Collapse
Affiliation(s)
- Jie Sun
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital, Sichuan University, Chengdu, China; Medical College, Tibet University, Lhasa, China
| | - Fan Yang
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Yanyi Zheng
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Chen Huang
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoli Fan
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital, Sichuan University, Chengdu, China.
| | - Li Yang
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
20
|
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
|
21
|
Maali Y, Flores Molina M, Khedr O, Abdelnabi MN, Dion J, Hassan GS, Shoukry NH. Two transcriptionally and functionally distinct waves of neutrophils during mouse acute liver injury. Hepatol Commun 2024; 8:e0459. [PMID: 38896080 PMCID: PMC11186811 DOI: 10.1097/hc9.0000000000000459] [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: 12/19/2023] [Accepted: 03/28/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Neutrophils are key mediators of inflammation during acute liver injury (ALI). Emerging evidence suggests that they also contribute to injury resolution and tissue repair. However, the different neutrophil subsets involved in these processes and their kinetics are undefined. Herein, we characterized neutrophil kinetics and heterogeneity during ALI. METHODS We used the carbon tetrachloride model of ALI and employed flow cytometry, tissue imaging, and quantitative RT-PCR to characterize intrahepatic neutrophils during the necroinflammatory early and late repair phases of the wound healing response to ALI. We FACS sorted intrahepatic neutrophils at key time points and examined their transcriptional profiles using RNA-sequencing. Finally, we evaluated neutrophil protein translation, mitochondrial function and metabolism, reactive oxygen species content, and neutrophil extracellular traps generation. RESULTS We detected 2 temporarily distinct waves of neutrophils during (1) necroinflammation (at 24 hours after injury) and (2) late repair (at 72 hours). Early neutrophils were proinflammatory, characterized by: (1) upregulation of inflammatory cytokines, (2) activation of the noncanonical NF-κB pathway, (3) reduction of protein translation, (4) decreased oxidative phosphorylation, and (5) higher propensity to generate reactive oxygen species and neutrophil extracellular traps. In contrast, late neutrophils were prorepair and enriched in genes and pathways associated with tissue repair and angiogenesis. Finally, early proinflammatory neutrophils were characterized by the expression of a short isoform of C-X-C chemokine receptor 5, while the late prorepair neutrophils were characterized by the expression of C-X-C chemokine receptor 4. CONCLUSIONS This study underscores the phenotypic and functional heterogeneity of neutrophils and their dual role in inflammation and tissue repair during ALI.
Collapse
Affiliation(s)
- Yousef Maali
- Immunopathology Axis, Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, Quebec, Canada
- Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, Quebec, Canada
| | - Manuel Flores Molina
- Immunopathology Axis, Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, Quebec, Canada
- Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, Quebec, Canada
| | - Omar Khedr
- Immunopathology Axis, Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, Quebec, Canada
| | - Mohamed N. Abdelnabi
- Immunopathology Axis, Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, Quebec, Canada
- Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, Quebec, Canada
| | - Jessica Dion
- Immunopathology Axis, Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, Quebec, Canada
| | - Ghada S. Hassan
- Immunopathology Axis, Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, Quebec, Canada
| | - Naglaa H. Shoukry
- Immunopathology Axis, Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, Quebec, Canada
- Departement de médecine, Université de Montréal, Montréal, Quebec, Canada
| |
Collapse
|
22
|
Roy A, Ghoshal UC, Goenka MK. Liver and Brain Disorders. CURRENT HEPATOLOGY REPORTS 2024; 23:404-413. [DOI: 10.1007/s11901-024-00668-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/15/2024] [Indexed: 01/04/2025]
|
23
|
Wu YP, Li FC, Ma HY, Yang XY, Zuo J, Tian YX, Lv L, Wang K, Fan YC. Characteristics and risk factors for invasive fungal infection in hospitalized patients with acute-on-chronic hepatitis B liver failure: a retrospective cohort study from 2010 to 2023. Front Microbiol 2024; 15:1391814. [PMID: 38601929 PMCID: PMC11004317 DOI: 10.3389/fmicb.2024.1391814] [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: 02/26/2024] [Accepted: 03/12/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND AND AIM The global burden of invasive fungal infections (IFIs) is emerging in immunologic deficiency status from various disease. Patients with acute-on-chronic hepatitis B liver failure (ACHBLF) are prone to IFI and their conditions are commonly exacerbated by IFI. However, little is known about the characteristics and risk factors for IFI in hospitalized ACHBLF patients. METHODS A total of 243 hospitalized ACHBLF patients were retrospectively enrolled from January 2010 to July 2023. We performed restricted cubic spline analysis to determine the non-linear associations between independent variables and IFI. The risk factors for IFI were identified using logistic regression and the extreme gradient boosting (XGBoost) algorithm. The effect values of the risk factors were determined by the SHapley Additive exPlanations (SHAP) method. RESULTS There were 24 ACHBLF patients (9.84%) who developed IFI on average 17.5 (13.50, 23.00) days after admission. The serum creatinine level showed a non-linear association with the possibility of IFI. Multiple logistic regression revealed that length of hospitalization (OR = 1.05, 95% CI: 1.02-1.08, P = 0.002) and neutrophilic granulocyte percentage (OR = 1.04, 95% CI: 1.00-1.09, P = 0.042) were independent risk factors for IFI. The XGBoost algorithm showed that the use of antibiotics (SHAP value = 0.446), length of hospitalization (SHAP value = 0.406) and log (qHBV DNA) (SHAP value = 0.206) were the top three independent risk factors for IFI. Furthermore, interaction analysis revealed no multiplicative effects between the use of antibiotics and the use of glucocorticoids (P = 0.990). CONCLUSION IFI is a rare complication that leads to high mortality in hospitalized ACHBLF patients, and a high neutrophilic granulocyte percentage and length of hospitalization are independent risk factors for the occurrence of IFI.
Collapse
Affiliation(s)
- Yin-Ping Wu
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
| | - Feng-Cai Li
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
| | - Hang-Yu Ma
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
| | - Xue-Yan Yang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
| | - Jing Zuo
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
| | - Yu-Xin Tian
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
| | - Li Lv
- Clinical Follow-up Center, Qilu Hospital of Shandong University, Jinan, China
| | - Kai Wang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
- Hepatology Institute of Shandong University, Jinan, China
| | - Yu-Chen Fan
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
- Hepatology Institute of Shandong University, Jinan, China
| |
Collapse
|
24
|
Zhu S, Yu Y, Hong Q, Li C, Zhang H, Guo K. Neutrophil Extracellular Traps Upregulate p21 and Suppress Cell Cycle Progression to Impair Endothelial Regeneration after Inflammatory Lung Injury. J Clin Med 2024; 13:1204. [PMID: 38592032 PMCID: PMC10931969 DOI: 10.3390/jcm13051204] [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: 12/08/2023] [Revised: 01/23/2024] [Accepted: 02/10/2024] [Indexed: 04/10/2024] Open
Abstract
Background: Sepsis is a major cause of ICU admissions, with high mortality and morbidity. The lungs are particularly vulnerable to infection and injury, and restoration of vascular endothelial homeostasis after injury is a crucial determinant of outcome. Neutrophil extracellular trap (NET) release strongly correlates with the severity of lung tissue damage. However, little is known about whether NETs affect endothelial cell (EC) regeneration and repair. Methods: Eight- to ten-week-old male C57BL/6 mice were injected intraperitoneally with a sublethal dose of LPS to induce acute lung inflammatory injury or with PBS as a control. Blood samples and lung tissues were collected to detect NET formation and lung endothelial cell proliferation. Human umbilical vein endothelial cells (HUVECs) were used to determine the role of NETs in cell cycle progression in vitro. Results: Increased NET formation and impaired endothelial cell proliferation were observed in mice with inflammatory lung injury following septic endotoxemia. Degradation of NETs with DNase I attenuated lung inflammation and facilitated endothelial regeneration. Mechanistically, NETs induced p21 upregulation and cell cycle stasis to impair endothelial repair. Conclusions: Our findings suggest that NET formation impairs endothelial regeneration and vascular repair through the induction of p21 and cell cycle arrest during inflammatory lung injury.
Collapse
Affiliation(s)
- Shuainan Zhu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (S.Z.); (Y.Y.); (Q.H.); (C.L.)
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai 210000, China
| | - Ying Yu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (S.Z.); (Y.Y.); (Q.H.); (C.L.)
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai 210000, China
| | - Qianya Hong
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (S.Z.); (Y.Y.); (Q.H.); (C.L.)
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai 210000, China
| | - Chenning Li
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (S.Z.); (Y.Y.); (Q.H.); (C.L.)
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai 210000, China
| | - Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (S.Z.); (Y.Y.); (Q.H.); (C.L.)
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai 210000, China
| | - Kefang Guo
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (S.Z.); (Y.Y.); (Q.H.); (C.L.)
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai 210000, China
| |
Collapse
|
25
|
Ma S, Xiao Y, Zhang X, Xu Y, Zhu K, Zhang K, Li X, Zhou H, Chen G, Guo X. Dietary exposure to polystyrene microplastics exacerbates liver damage in fulminant hepatic failure via ROS production and neutrophil extracellular trap formation. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 907:167403. [PMID: 37820799 DOI: 10.1016/j.scitotenv.2023.167403] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/25/2023] [Accepted: 09/25/2023] [Indexed: 10/13/2023]
Abstract
With the increasing influx of microplastics (MPs) into the environment, their potential toxicity represents an increasing threat to human health. However, there is a lack of relevant research surrounding the biological toxicity associated with pre-exposure to MPs under pathological conditions. To fill this gap, we established a mouse model of fulminant hepatic failure after 14 days of pre-exposure to polystyrene (PS) MPs and investigated its biological response process under combined stimulation with lipopolysaccharide (LPS)/d-galactosamine (d-GalN) and PS-MPs. The results indicated that the stress response from exposure to PS-MPs exacerbated the death induced by LPS/d-GalN and reinforced the potential of liver damage in mice. The dominant roles of inflammation promotion, reactive oxygen species (ROS), and neutrophil extracellular traps in this process were confirmed by cellular reactive oxygen species assays and experiments on oxidative stress and inflammatory responses in the liver. Transcriptomic analysis revealed that PS-MPs exacerbated the expression levels of neutrophil extracellular traps in mice treated with LPS/d-GalN, and weakened the expression of genes involved in pathways related to peroxisome, taurine, and hypotaurine metabolism, which was further validated by reverse-transcription quantitative polymerase chain reaction. This study addresses the knowledge gap regarding the adverse effects caused by a pathological state upon exposure to MPs and provides a theoretical reference for further assessment of the underlying health risks of MPs.
Collapse
Affiliation(s)
- Sirui Ma
- College of Natural Resources and Environment, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yao Xiao
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Beijing 100850, China
| | - Xiaoyong Zhang
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Beijing 100850, China
| | - Yibo Xu
- College of Natural Resources and Environment, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Kai Zhu
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Beijing 100850, China
| | - Kexin Zhang
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Beijing 100850, China; Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Park Road 977, Yanji, Jilin 133002, China
| | - Xinze Li
- Minkang Community Health Service Center, Changchun, Jilin 130041, China
| | - Hong Zhou
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Beijing 100850, China.
| | - Gan Chen
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Beijing 100850, China.
| | - Xuetao Guo
- College of Natural Resources and Environment, Northwest A&F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
26
|
Wang Y, Shi C, Guo J, Zhang D, Zhang Y, Zhang L, Gong Z. IDH1/MDH1 deacetylation promotes acute liver failure by regulating NETosis. Cell Mol Biol Lett 2024; 29:8. [PMID: 38172700 PMCID: PMC10765752 DOI: 10.1186/s11658-023-00529-7] [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: 08/03/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Acute liver failure (ALF) is a life-threatening disease, but its pathogenesis is not fully understood. NETosis is a novel mode of cell death. Although the formation of neutrophil extracellular traps (NETs) has been found in various liver diseases, the specific mechanism by which NETosis regulates the development of ALF is unclear. In this article, we explore the role and mechanism of NETosis in the pathogenesis of ALF. METHODS Clinically, we evaluated NETs-related markers in the liver and peripheral neutrophils of patients with ALF. In in vitro experiments, HL-60 cells were first induced to differentiate into neutrophil-like cells (dHL-60 cells) with dimethyl sulfoxide (DMSO). NETs were formed by inducing dHL-60 cells with PMA. In in vivo experiments, the ALF model in mice was established with LPS/D-gal, and the release of NETs was detected by immunofluorescence staining and western blotting. Finally, the acetylation levels of IDH1 and MDH1 were detected in dHL-60 cells and liver samples by immunoprecipitation. RESULTS Clinically, increased release of NETs in liver tissue was observed in patients with ALF, and NETs formation was detected in neutrophils from patients with liver failure. In dHL-60 cells, mutations at IDH1-K93 and MDH1-K118 deacetylate IDH1 and MDH1, which promotes the formation of NETs. In a mouse model of ALF, deacetylation of IDH1 and MDH1 resulted in NETosis and promoted the progression of acute liver failure. CONCLUSIONS Deacetylation of IDH1 and MDH1 reduces their activity and promotes the formation of NETs. This change aggravates the progression of acute liver failure.
Collapse
Affiliation(s)
- Yukun Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Chunxia Shi
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Jin Guo
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Danmei Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Yanqiong Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Long Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Zuojiong Gong
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China.
| |
Collapse
|
27
|
Schulte A, Groeneveld DJ, Wei Z, Hazel B, Bernard MP, Poole LG, Luyendyk JP. Neutrophil-dependent hepatic platelet accumulation and liver injury revealed by acetaminophen dose-response studies. Res Pract Thromb Haemost 2024; 8:102323. [PMID: 38404941 PMCID: PMC10883821 DOI: 10.1016/j.rpth.2024.102323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 02/27/2024] Open
Abstract
Background Acetaminophen (APAP) overdose is a leading cause of drug-induced acute liver failure (ALF). Neutrophil activation has been associated with poor outcomes in patients with ALF and is proposed to amplify coagulation in this context. However, the precise role of neutrophils in APAP-induced liver injury is not known. Methods We used a dual antibody-mediated neutrophil depletion strategy to determine the role of neutrophils in mice challenged with different doses of APAP (300 or 600 mg/kg) that produce hepatotoxicity and ALF-like pathology. Results Flow cytometry confirmed depletion of neutrophils in whole blood prior to APAP challenge. Mice given isotype control and challenged with 300 mg/kg APAP developed marked hepatocellular necrosis and showed an increase in biomarkers of coagulation cascade activation. Neutrophil depletion (anti-Ly6G) did not affect either liver injury or coagulation activation in mice challenged with 300 mg/kg APAP. Mice given isotype control and challenged with 600 mg/kg APAP developed hepatic necrosis alongside marked hemorrhage and congestion indicative of vascular injury. Interestingly, hepatic neutrophil and platelet accumulation were increased in mice given 600 mg/kg APAP compared with those given the lower APAP dose. Neutrophil depletion significantly reduced the severity of liver necrosis in mice challenged with 600 mg/kg APAP, without significantly impacting biomarkers of coagulation activity. Notably, neutrophil depletion significantly reduced hepatic platelet accumulation in mice challenged with 600 mg/kg APAP. Conclusion The results indicate a role of neutrophils in APAP-induced liver injury that is dependent on the APAP dose and suggest involvement of neutrophil-platelet interactions in promoting hepatic injury in experimental APAP-induced ALF.
Collapse
Affiliation(s)
- Anthony Schulte
- Department of Pathobiology & Diagnostic Investigation, Michigan State University, East Lansing, Michigan, USA
| | - Dafna J. Groeneveld
- Department of Pathobiology & Diagnostic Investigation, Michigan State University, East Lansing, Michigan, USA
| | - Zimu Wei
- Department of Pathobiology & Diagnostic Investigation, Michigan State University, East Lansing, Michigan, USA
| | - Bianca Hazel
- Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| | - Matthew P. Bernard
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Lauren G. Poole
- Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| | - James P. Luyendyk
- Department of Pathobiology & Diagnostic Investigation, Michigan State University, East Lansing, Michigan, USA
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
28
|
Brunnthaler L, Pereyra D, Brenner M, Santol J, Herrmann L, Schrottmaier WC, Pirabe A, Schmuckenschlager A, Kim S, Kern AE, Huber FX, Michels LE, Brostjan C, Salzmann M, Hohensinner P, Kain R, Gruenberger T, Starlinger P, Assinger A. Intrahepatic neutrophil accumulation and extracellular trap formation are associated with posthepatectomy liver failure. Hepatol Commun 2024; 8:e0348. [PMID: 38099865 PMCID: PMC10727591 DOI: 10.1097/hc9.0000000000000348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 10/13/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Posthepatectomy liver failure (PHLF) represents a life-threatening complication with limited therapeutic options. Neutrophils play a critical and dynamic role during regeneratory processes, but their role in human liver regeneration is incompletely understood, especially as underlying liver disease, detectable in the majority of patients, critically affects hepatic regeneration. Here we explored intrahepatic neutrophil accumulation and neutrophil extracellular traps (NETs) in patients with PHLF and validated the functional relevance of NETs in a murine partial hepatectomy (PHx) model. METHODS We investigated the influx of neutrophils, macrophages, eosinophils, and mast cells and the presence of their respective extracellular traps in liver biopsies of 35 patients undergoing hepatectomy (10 patients with PHLF) before and after the initiation of liver regeneration by fluorescence microscopy. In addition, NET formation and neutrophil activation were confirmed by plasma analysis of 99 patients (24 patients with PHLF) before and up to 5 days after surgery. Furthermore, we inhibited NETs via DNase I in a murine PHx model of mice with metabolically induced liver disease. RESULTS We detected rapid intrahepatic neutrophil accumulation, elevated levels of myeloperoxidase release, and NET formation in regenerating human livers, with a significantly higher increase of infiltrating neutrophils and NETs in patients with PHLF. Circulating markers of neutrophil activation, including elastase, myeloperoxidase, and citrullinated histone H3, correlated with markers of liver injury. In a murine PHx model, we showed that the inhibition of NET accelerated hepatocyte proliferation and liver regeneration. CONCLUSIONS Patients with PHLF showed accelerated intrahepatic neutrophil infiltration and NET formation, which were associated with liver damage. Further, we identified postsurgical myeloperoxidase levels as predictive markers for adverse outcomes and observed that blocking NETs in a murine PHx model accelerated tissue regeneration.
Collapse
Affiliation(s)
- Laura Brunnthaler
- Department of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - David Pereyra
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, General Hospital, Vienna, Austria
| | - Miriam Brenner
- Department of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Jonas Santol
- Department of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
- Department of Surgery, HPB Center, Viennese Health Network, Clinic Favoriten and Sigmund Freud Private University, Vienna, Austria
| | - Lukas Herrmann
- Department of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Waltraud C. Schrottmaier
- Department of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Anita Pirabe
- Department of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Anna Schmuckenschlager
- Department of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Sarang Kim
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, General Hospital, Vienna, Austria
| | - Anna Emilia Kern
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, General Hospital, Vienna, Austria
| | - Felix Xaver Huber
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, General Hospital, Vienna, Austria
| | - Lisa Emilie Michels
- Department of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Christine Brostjan
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, General Hospital, Vienna, Austria
| | - Manuel Salzmann
- Department of Medicine II, Division of Cardiology, Medical University of Vienna, General Hospital, Vienna, Austria
| | - Philipp Hohensinner
- Center for Biomedical Research, Division of Biomedical Research, Medical University of Vienna, Vienna, Austria
| | - Renate Kain
- Department of Pathology, Medical University of Vienna, General Hospital, Vienna, Austria
| | - Thomas Gruenberger
- Department of Surgery, HPB Center, Viennese Health Network, Clinic Favoriten and Sigmund Freud Private University, Vienna, Austria
| | - Patrick Starlinger
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Alice Assinger
- Department of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
29
|
Wang Z, Zhang C, Meng J, Jiao Z, Bao W, Tian H, Wu C, Chai W, Li R, Liu Z, Ma G, Mei X, Wei W. A Targeted Exosome Therapeutic Confers Both CfDNA Scavenging and Macrophage Polarization for Ameliorating Rheumatoid Arthritis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2302503. [PMID: 37681753 DOI: 10.1002/adma.202302503] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 08/20/2023] [Indexed: 09/09/2023]
Abstract
Only a minority of rheumatoid arthritis (RA) patients achieve disease remission, so the exploration of additional pathogenic factors and the development of new therapeutics are needed. Here, strong correlations among the cell-free DNA (cfDNA) level and the inflammatory response in clinical synovial fluid samples and RA disease activity are discovered. The important role of cfDNA in disease development in a collagen-induced arthritis (CIA) murine model is also demonstrated. Building on these findings, a novel therapeutic based on anti-inflammatory (M2) macrophage-derived exosomes as chassis, that are modified with both oligolysine and matrix metalloproteinase (MMP)-cleavable polyethylene glycol (PEG) on the membrane, is developed. After intravenous injection, PEG-enabled prolonged circulation and C─C motif chemokine ligand-directed accumulation together result in enrichment at inflamed joints. Following subsequent MMP cleavage, the positively charged oligolysine is exposed for cfDNA scavenging, while exosomes induce M2 polarization. By using a classical CIA murine model and a newly established CIA canine model, it is demonstrated that the rationally designed exosome therapeutic substantially suppresses inflammation in joints and provides strong chondroprotection and osteoprotection, revealing its potential for effective CIA amelioration.
Collapse
Affiliation(s)
- Zhe Wang
- Department of Orthopedics, Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121002, P. R. China
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- Key Laboratory of Medical Tissue Engineering of Liaoning, Jinzhou Medical University, Jinzhou, 121001, P. R. China
| | - Chuanjie Zhang
- Department of Orthopedics, Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121002, P. R. China
- Key Laboratory of Medical Tissue Engineering of Liaoning, Jinzhou Medical University, Jinzhou, 121001, P. R. China
| | - Jiaqi Meng
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Zhouguang Jiao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Weier Bao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - He Tian
- Key Laboratory of Medical Tissue Engineering of Liaoning, Jinzhou Medical University, Jinzhou, 121001, P. R. China
| | - Chao Wu
- Key Laboratory of Medical Tissue Engineering of Liaoning, Jinzhou Medical University, Jinzhou, 121001, P. R. China
| | - Wei Chai
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing, 100037, P. R. China
| | - Rui Li
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing, 100037, P. R. China
| | - Zheng Liu
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing, 100037, P. R. China
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xifan Mei
- Department of Orthopedics, Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121002, P. R. China
- Key Laboratory of Medical Tissue Engineering of Liaoning, Jinzhou Medical University, Jinzhou, 121001, P. R. China
| | - Wei Wei
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
30
|
Tang C, Cen L, Zeng H, Zhang X, Liu P, Chen Y, Song X, Lin B, Zhang X, Yu C, Xu C. Inhibiting Hepatocyte Uric Acid Synthesis and Reabsorption Ameliorates Acetaminophen-Induced Acute Liver Injury in Mice. Cell Mol Gastroenterol Hepatol 2023; 17:251-265. [PMID: 37879407 PMCID: PMC10765060 DOI: 10.1016/j.jcmgh.2023.10.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 10/17/2023] [Accepted: 10/17/2023] [Indexed: 10/27/2023]
Abstract
BACKGROUND & AIMS Acetaminophen (APAP) overdose is the most common cause of drug-induced liver injury worldwide. Uric acid (UA) is involved in sterile inflammation in many organs, but its role in APAP-induced liver injury remains elusive. METHODS We quantified the concentration of UA in the serum and liver tissues of APAP-overdosed mice and explored the changes in proteins involved in UA synthesis, absorption, and degeneration on APAP stimulation. We also examined the effects of inhibiting hepatocyte UA synthesis or reabsorption on APAP-induced liver injury in mice. Furthermore, we explored the process of UA clearance by peripheral macrophages. RESULTS APAP overdose significantly increased intrahepatic UA contents, which occurred earlier than apparent hepatocyte injury in APAP-overdosed mice. APAP overdose induced significant DNA leakage and may thereby increase the substrate of UA synthesis. APAP overdose also significantly increased the enzymatic activity of xanthine oxidase and urate oxidase and decreased the expression of the UA reabsorption transporter GLUT9 in hepatocytes. Inhibiting hepatocyte UA synthesis by febuxostat or reabsorption by hepatic-specific knockout of GLUT9 alleviated APAP-induced liver injury. Further experiments showed that monosodium urate but not soluble UA may be a major form of UA mediating hepatocyte injury. Additionally, monosodium urate further recruited circulating macrophages into the liver and then aggravated inflammation by increasing the levels of inflammatory factors and reactive oxygen species. Deletion of macrophages significantly ameliorated APAP-induced liver injury in mice. CONCLUSIONS APAP overdose induces excessive UA production and leads to local high concentrations in the liver, which further injures cells and induces liver inflammation. Inhibiting the production of UA may be a potential therapeutic option for treating APAP-induced liver injury.
Collapse
Affiliation(s)
- Chenxi Tang
- Department of Gastroenterology, Zhejiang Provincial Clinical Research Center for Digestive Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Li Cen
- Department of Gastroenterology, Zhejiang Provincial Clinical Research Center for Digestive Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hang Zeng
- Department of Gastroenterology, Zhejiang Provincial Clinical Research Center for Digestive Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaofen Zhang
- Department of Gastroenterology, Zhejiang Provincial Clinical Research Center for Digestive Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Peihao Liu
- Department of Gastroenterology, Zhejiang Provincial Clinical Research Center for Digestive Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yishu Chen
- Department of Gastroenterology, Zhejiang Provincial Clinical Research Center for Digestive Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xin Song
- Department of Gastroenterology, Zhejiang Provincial Clinical Research Center for Digestive Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bingru Lin
- Department of Gastroenterology, Zhejiang Provincial Clinical Research Center for Digestive Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xuequn Zhang
- Department of Gastroenterology, Zhejiang Provincial Clinical Research Center for Digestive Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chaohui Yu
- Department of Gastroenterology, Zhejiang Provincial Clinical Research Center for Digestive Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Chengfu Xu
- Department of Gastroenterology, Zhejiang Provincial Clinical Research Center for Digestive Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
31
|
Li C, Wu J, Zhang L, Wang F, Xu L, Zhao Y, Xiao Y, Zhuang F, Hou L, Zhao D, She Y, Xie D, Chen C. Brief Report: Acetaminophen Reduces Neoadjuvant Chemoimmunotherapy Efficacy in Patients With NSCLC by Promoting Neutrophil Extracellular Trap Formation: Analysis From a Phase 2 Clinical Trial. JTO Clin Res Rep 2023; 4:100556. [PMID: 37654895 PMCID: PMC10466912 DOI: 10.1016/j.jtocrr.2023.100556] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/26/2023] [Accepted: 07/15/2023] [Indexed: 09/02/2023] Open
Abstract
Introduction Neoadjuvant chemoimmunotherapy has recently been the standard of care for resectable locally advanced NSCLC. Factors affecting the neoadjuvant immunotherapy efficacy, however, remain elusive. Metabolites have been found to modulate immunity and associate with immunotherapeutic efficacy in advanced tumors. Therefore, we aimed to investigate the impact of plasma metabolites on the pathologic response after neoadjuvant chemoimmunotherapy. Methods Patients with stage IIIA (N2) NSCLC who underwent neoadjuvant chemoimmunotherapy in a prospective phase 2 clinical trial (NCT04422392) were enrolled. Metabolomic profiling of the plasma before treatment was performed using liquid chromatography-mass spectrometry. A Lewis lung carcinoma mouse model was further used to investigate the underlying mechanisms. Proteomics and multiplexed immunofluorescence of the mice tumor were performed. Results A total of 39 patients who underwent three cycles of anti-programmed cell death-protein 1 (anti-PD-1) (sintilimab) and chemotherapy were included. The level of acetaminophen (APAP) was found to be significantly elevated in patients who did not achieve major pathologic response. The level of APAP remained an independent predictor for major pathologic response in multivariate logistic analysis. In the Lewis lung carcinoma mouse model, combination of APAP and anti-PD-1 treatment significantly reduced the treatment efficacy compared with anti-PD-1 treatment alone. Proteomics of the tumor revealed that myeloid leukocyte activation and neutrophil activation pathways were enriched after APAP treatment. Tumor microenvironment featuring analysis also revealed that the combination treatment group was characterized with more abundant neutrophil signature. Further multiplexed immunofluorescence confirmed that more neutrophil extracellular trap formation was observed in the combination treatment group. Conclusions APAP could impair neoadjuvant chemoimmunotherapy efficacy in patients with NSCLC by promoting neutrophil activation and neutrophil extracellular trap formation.
Collapse
Affiliation(s)
- Chongwu Li
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Junqi Wu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Lei Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Fang Wang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Long Xu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Yue Zhao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Yun Xiao
- DeepKinase Co, Ltd., Beijing, People’s Republic of China
| | - Fenghui Zhuang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Likun Hou
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Deping Zhao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Yunlang She
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Dong Xie
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| |
Collapse
|
32
|
Sun M, Chen P, Xiao K, Zhu X, Zhao Z, Guo C, He X, Shi T, Zhong Q, Jia Y, Tao Y, Li M, Leong KW, Shao D. Circulating Cell-Free DNAs as a Biomarker and Therapeutic Target for Acetaminophen-Induced Liver Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206789. [PMID: 37035952 PMCID: PMC10238175 DOI: 10.1002/advs.202206789] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 01/08/2023] [Indexed: 06/04/2023]
Abstract
Acetaminophen (APAP) overdose is a leading cause of drug-induced liver injury and acute liver failure, while the detection, prognosis prediction, and therapy for APAP-induced liver injury (AILI) remain improved. Here, it is determined that the temporal pattern of circulating cell-free DNA (cfDNA) is strongly associated with damage and inflammation parameters in AILI. CfDNA is comparable to alanine aminotransferase (ALT) in predicting mortality and outperformed ALT when combined with ALT in AILI. The depletion of cfDNA or neutrophils alleviates liver damage, while the addition of cfDNA or adoptive transfer of neutrophils exacerbates the damage. The combination of DNase I and N-acetylcysteine attenuates AILI significantly. This study establishes that cfDNA is a mechanistic biomarker to predict mortality in AILI mice. The combination of scavenging cfDNA and reducing oxidative damage provides a promising treatment for AILI.
Collapse
Affiliation(s)
- Madi Sun
- School of Biomedical Sciences and EngineeringSouth China University of TechnologyGuangzhou International CampusGuangzhouGuangdong510630China
- National Engineering Research Center for Tissue Restoration and ReconstructionSouth China University of TechnologyGuangzhou International CampusGuangzhouGuangdong510630China
| | - Peiyu Chen
- School of Biomedical Sciences and EngineeringSouth China University of TechnologyGuangzhou International CampusGuangzhouGuangdong510630China
- National Engineering Research Center for Tissue Restoration and ReconstructionSouth China University of TechnologyGuangzhou International CampusGuangzhouGuangdong510630China
| | - Kai Xiao
- National Engineering Research Center for Tissue Restoration and ReconstructionSouth China University of TechnologyGuangzhou International CampusGuangzhouGuangdong510630China
- School of MedicineSouth China University of TechnologyGuangzhou International CampusGuangzhouGuangdong510006China
| | - Xiang Zhu
- Laboratory of Biomaterials and Translational MedicineThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510006China
| | - Zhibin Zhao
- School of MedicineSouth China University of TechnologyGuangzhou International CampusGuangzhouGuangdong510006China
| | - Chenyang Guo
- School of Biomedical Sciences and EngineeringSouth China University of TechnologyGuangzhou International CampusGuangzhouGuangdong510630China
- National Engineering Research Center for Tissue Restoration and ReconstructionSouth China University of TechnologyGuangzhou International CampusGuangzhouGuangdong510630China
| | - Xuan He
- School of Biomedical Sciences and EngineeringSouth China University of TechnologyGuangzhou International CampusGuangzhouGuangdong510630China
- National Engineering Research Center for Tissue Restoration and ReconstructionSouth China University of TechnologyGuangzhou International CampusGuangzhouGuangdong510630China
| | - Tongfei Shi
- School of Biomedical Sciences and EngineeringSouth China University of TechnologyGuangzhou International CampusGuangzhouGuangdong510630China
- National Engineering Research Center for Tissue Restoration and ReconstructionSouth China University of TechnologyGuangzhou International CampusGuangzhouGuangdong510630China
| | - Qingguo Zhong
- Laboratory of Biomaterials and Translational MedicineThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510006China
| | - Yong Jia
- School of NursingJilin UniversityChangchunJilin130021China
| | - Yu Tao
- Laboratory of Biomaterials and Translational MedicineThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510006China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational MedicineThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510006China
| | - Kam W. Leong
- Department of Systems BiologyColumbia UniversityNew YorkNY10032USA
| | - Dan Shao
- School of Biomedical Sciences and EngineeringSouth China University of TechnologyGuangzhou International CampusGuangzhouGuangdong510630China
- National Engineering Research Center for Tissue Restoration and ReconstructionSouth China University of TechnologyGuangzhou International CampusGuangzhouGuangdong510630China
- Guangdong Provincial Key Laboratory of Biomedical EngineeringKey Laboratory of Biomedical Materials and Engineering of the Ministry of EducationSouth China University of TechnologyGuangzhouGuangdong510006China
| |
Collapse
|
33
|
Chen L, Yuan L, Yang J, Pan Y, Wang H. Identification of key immune-related genes associated with LPS/D-GalN-induced acute liver failure in mice based on transcriptome sequencing. PeerJ 2023; 11:e15241. [PMID: 37168540 PMCID: PMC10166078 DOI: 10.7717/peerj.15241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 03/28/2023] [Indexed: 05/13/2023] Open
Abstract
BACKGROUND The aim of this study was to identify key immune-related genes in acute liver failure (ALF) by constructing an ALF mouse model for transcriptome sequencing. METHODS The C57BL/6 mouse with ALF model was induced by lipopolysaccharide (LPS)/ D-galactosamine (D-GalN). After successful modelling, the liver tissues of all mice were obtained for transcriptome sequencing. The key immune-related genes in mice with ALF were identified by differential expression analysis, immune infiltration analysis, weighted gene co-expression network analysis (WGCNA), enrichment analysis, and protein-protein interaction (PPI) analysis. RESULTS An LPS/D-GalN-induced ALF mouse model was successfully constructed, and transcriptome sequencing was performed. Significant differences in the proportions of monocytes, macrophages M0, macrophages M1 and neutrophils were shown by immune infiltration analysis, and 5255 genes highly associated with these four immune cells were identified by WGCNA. These immune genes were found to be significantly enriched in the TNF signalling pathway by enrichment analysis. Finally, PPI analysis was performed on genes enriched in this pathway and three key genes (CXCL1, CXCL10 and IL1B) were screened out and revealed to be significantly upregulated in ALF. CONCLUSIONS Key immune-related genes in ALF were identified in this study, which may provide not only potential therapeutic targets for treating ALF and improving its prognosis, but also a reliable scientific basis for the immunotherapy of the disease.
Collapse
Affiliation(s)
- Ling Chen
- Department of Infectious Disease, Zhejiang Hospital, Hangzhou, China
| | - Li Yuan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jingle Yang
- Department of Infectious Disease, Zhejiang Hospital, Hangzhou, China
| | - Yizhi Pan
- Department of Infectious Disease, Zhejiang Hospital, Hangzhou, China
| | - Hong Wang
- Department of Infectious Disease, Zhejiang Hospital, Hangzhou, China
| |
Collapse
|
34
|
Kyritsi K, Wu N, Zhou T, Carpino G, Baiocchi L, Kennedy L, Chen L, Ceci L, Meyer AA, Barupala N, Franchitto A, Onori P, Ekser B, Gaudio E, Wu C, Marakovits C, Chakraborty S, Francis H, Glaser S, Alpini G. Knockout of secretin ameliorates biliary and liver phenotypes during alcohol-induced hepatotoxicity. Cell Biosci 2023; 13:5. [PMID: 36624475 PMCID: PMC9830859 DOI: 10.1186/s13578-022-00945-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Alcohol-related liver disease (ALD) is characterized by ductular reaction (DR), liver inflammation, steatosis, fibrosis, and cirrhosis. The secretin (Sct)/secretin receptor (SR) axis (expressed only by cholangiocytes) regulates liver phenotypes in cholestasis. We evaluated the role of Sct signaling on ALD phenotypes. METHODS We used male wild-type and Sct-/- mice fed a control diet (CD) or ethanol (EtOH) for 8 wk. Changes in liver phenotypes were measured in mice, female/male healthy controls, and patients with alcoholic cirrhosis. Since Cyp4a10 and Cyp4a11/22 regulate EtOH liver metabolism, we measured their expression in mouse/human liver. We evaluated: (i) the immunoreactivity of the lipogenesis enzyme elongation of very-long-chain fatty acids 1 (Elovl, mainly expressed by hepatocytes) in mouse/human liver sections by immunostaining; (ii) the expression of miR-125b (that is downregulated in cholestasis by Sct) in mouse liver by qPCR; and (iii) total bile acid (BA) levels in mouse liver by enzymatic assay, and the mRNA expression of genes regulating BA synthesis (cholesterol 7a-hydroxylase, Cyp27a1, 12a-hydroxylase, Cyp8b1, and oxysterol 7a-hydroxylase, Cyp7b11) and transport (bile salt export pump, Bsep, Na+-taurocholate cotransporting polypeptide, NTCP, and the organic solute transporter alpha (OSTa) in mouse liver by qPCR. RESULTS In EtOH-fed WT mice there was increased biliary and liver damage compared to control mice, but decreased miR-125b expression, phenotypes that were blunted in EtOH-fed Sct-/- mice. The expression of Cyp4a10 increased in cholangiocytes and hepatocytes from EtOH-fed WT compared to control mice but decreased in EtOH-fed Sct-/- mice. There was increased immunoreactivity of Cyp4a11/22 in patients with alcoholic cirrhosis compared to controls. The expression of miR-125b decreased in EtOH-fed WT mice but returned at normal values in EtOH-fed Sct-/- mice. Elovl1 immunoreactivity increased in patients with alcoholic cirrhosis compared to controls. There was no difference in BA levels between WT mice fed CD or EtOH; BA levels decreased in EtOH-fed Sct-/- compared to EtOH-fed WT mice. There was increased expression of Cyp27a1, Cyp8b1, Cyp7b1, Bsep, NTCP and Osta in total liver from EtOH-fed WT compared to control mice, which decreased in EtOH-fed Sct-/- compared to EtOH-fed WT mice. CONCLUSIONS Targeting Sct/SR signaling may be important for modulating ALD phenotypes.
Collapse
Affiliation(s)
- Konstantina Kyritsi
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nan Wu
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Tianhao Zhou
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Guido Carpino
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, La Sapienza University of Rome, Rome, Italy
| | | | - Lindsey Kennedy
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Division of Research, Indiana Center for Liver Research, Gastroenterology, Medicine, Richard L. Roudebush VA Medical Center and Indiana University, 702 Rotary Circle, Rm. 013C, Indianapolis, IN, 46202-2859, USA
| | - Lixian Chen
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ludovica Ceci
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, La Sapienza University of Rome, Rome, Italy
| | - Alison Ann Meyer
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nipuni Barupala
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Antonio Franchitto
- Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", Rome, Italy
| | - Paolo Onori
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, La Sapienza University of Rome, Rome, Italy
| | - Burcin Ekser
- Division of Transplant Surgery, Department of Surgery, Indiana University, Indianapolis, IN, USA
| | - Eugenio Gaudio
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, La Sapienza University of Rome, Rome, Italy
| | - Chaodong Wu
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Corinn Marakovits
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sanjukta Chakraborty
- Department of Medical Physiology, Texas A&M University School of Medicine, 8447 Riverside Parkway, MREB II, Room 2342, Bryan, TX, 77807-3260, USA
| | - Heather Francis
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.
- Division of Research, Indiana Center for Liver Research, Gastroenterology, Medicine, Richard L. Roudebush VA Medical Center and Indiana University, 702 Rotary Circle, Rm. 013C, Indianapolis, IN, 46202-2859, USA.
| | - Shannon Glaser
- Department of Medical Physiology, Texas A&M University School of Medicine, 8447 Riverside Parkway, MREB II, Room 2342, Bryan, TX, 77807-3260, USA.
| | - Gianfranco Alpini
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.
- Division of Research, Indiana Center for Liver Research, Gastroenterology, Medicine, Richard L. Roudebush VA Medical Center and Indiana University, 702 Rotary Circle, Rm. 013C, Indianapolis, IN, 46202-2859, USA.
| |
Collapse
|
35
|
Zhao Z, Pan Z, Zhang S, Ma G, Zhang W, Song J, Wang Y, Kong L, Du G. Neutrophil extracellular traps: A novel target for the treatment of stroke. Pharmacol Ther 2023; 241:108328. [PMID: 36481433 DOI: 10.1016/j.pharmthera.2022.108328] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/30/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
Stroke is a threatening cerebrovascular disease caused by thrombus with high morbidity and mortality rates. Neutrophils are the first to be recruited in the brain after stroke, which aggravate brain injury through multiple mechanisms. Neutrophil extracellular traps (NETs), as a novel regulatory mechanism of neutrophils, can trap bacteria and secret antimicrobial molecules, thereby degrading pathogenic factors and killing bacteria. However, NETs also exacerbate certain non-infectious diseases by activating autoimmune or inflammatory responses. NETs have been found to play important roles in the pathological process of stroke in recent years. In this review, the mechanisms of NETs formation, the physiological roles of NETs, and the dynamic changes of NETs after stroke are summarized. NETs participate in stroke through various mechanisms. NETs promote the coagulation cascade and interact with platelets to induce thrombosis. tPA induces the degranulation of neutrophils to form NETs, leading to hemorrhagic transformation and thrombolytic resistance. NETs aggravate stroke by mediating inflammation, atherosclerosis and vascular injury. In addition, the regulation of NETs in stroke, the potential of NETs as biomarker and the treatment of stroke targeting NETs are discussed. The increasing evidences suggest that NETs may be a potential target for stroke treatment. Inhibition of NETs formation or promotion of NETs degradation plays protective effects in stroke. However, how to avoid the adverse effects of NETs-targeted therapy deserves further study. In summary, this review provides a reference for the pathogenesis, drug targets, biomarkers and drug development of NETs in stroke.
Collapse
Affiliation(s)
- Ziyuan Zhao
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Zirong Pan
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Sen Zhang
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Guodong Ma
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Wen Zhang
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Junke Song
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Yuehua Wang
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Linglei Kong
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China.
| | - Guanhua Du
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
36
|
Liu Y, Yan P, Bin Y, Qin X, Wu Z. Neutrophil extracellular traps and complications of liver transplantation. Front Immunol 2022; 13:1054753. [PMID: 36466888 PMCID: PMC9712194 DOI: 10.3389/fimmu.2022.1054753] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 10/26/2022] [Indexed: 08/29/2023] Open
Abstract
Many end-stage liver disease etiologies are attributed to robust inflammatory cell recruitment. Neutrophils play an important role in inflammatory infiltration and neutrophil phagocytosis, oxidative burst, and degranulation. It has also been suggested that neutrophils may release neutrophil extracellular traps (NETs) to kill pathogens. It has been proven that neutrophil infiltration within the liver contributes to an inflammatory microenvironment and immune cell activation. Growing evidence implies that NETs are involved in the progression of numerous complications of liver transplantation, including ischemia-reperfusion injury, acute rejection, thrombosis, and hepatocellular carcinoma recurrence. NETs are discussed in this comprehensive review, focusing on their effects on liver transplantation complications. Furthermore, we discuss NETs as potential targets for liver transplantation therapy.
Collapse
Affiliation(s)
- Yanyao Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ping Yan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yue Bin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoyan Qin
- Department of General Surgery and Trauma Surgery, Children’s Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Zhongjun Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
37
|
Morris SM, Chauhan A. The role of platelet mediated thromboinflammation in acute liver injury. Front Immunol 2022; 13:1037645. [PMID: 36389830 PMCID: PMC9647048 DOI: 10.3389/fimmu.2022.1037645] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/14/2022] [Indexed: 12/03/2022] Open
Abstract
Acute liver injuries have wide and varied etiologies and they occur both in patients with and without pre-existent chronic liver disease. Whilst the pathophysiological mechanisms remain distinct, both acute and acute-on-chronic liver injury is typified by deranged serum transaminase levels and if severe or persistent can result in liver failure manifest by a combination of jaundice, coagulopathy and encephalopathy. It is well established that platelets exhibit diverse functions as immune cells and are active participants in inflammation through processes including immunothrombosis or thromboinflammation. Growing evidence suggests platelets play a dualistic role in liver inflammation, shaping the immune response through direct interactions and release of soluble mediators modulating function of liver sinusoidal endothelial cells, stromal cells as well as migrating and tissue-resident leucocytes. Elucidating the pathways involved in initiation, propagation and resolution of the immune response are of interest to identify therapeutic targets. In this review the provocative role of platelets is outlined, highlighting beneficial and detrimental effects in a spatial, temporal and disease-specific manner.
Collapse
Affiliation(s)
- Sean M. Morris
- The Liver Unit, University Hospitals Birmingham, Birmingham, United Kingdom
| | - Abhishek Chauhan
- The Liver Unit, University Hospitals Birmingham, Birmingham, United Kingdom
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- *Correspondence: Abhishek Chauhan,
| |
Collapse
|
38
|
Meng XY, Lu QY, Zhang JF, Li JF, Shi MY, Huang SY, Yu SF, Zhao YM, Fan HJ. A novel animal model of primary blast lung injury and its pathological changes in mice. J Trauma Acute Care Surg 2022; 93:530-537. [PMID: 35261371 PMCID: PMC9488943 DOI: 10.1097/ta.0000000000003571] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND Primary blast lung injury (PBLI) is a major cause of death in military conflict and terrorist attacks on civilian populations. However, the mechanisms of PBLI are not well understood, and a standardized animal model is urgently needed. This study aimed to establish an animal model of PBLI for laboratory study. METHODS The animal model of PBLI was established using a self-made mini shock tube simulation device. In brief, mice were randomly divided into two groups: the control group and the model group, the model group were suffered 0.5 bar shock pressures. Mice were sacrificed at 2 hours, 4 hours, 6 hours, 12 hours, and 24 hours after injury. Lung tissue gross observation, hematoxylin and eosin staining and lung pathology scoring were performed to evaluated lung tissue damage. Evans blue dye leakage and bronchoalveolar lavage fluid examination were performed to evaluated pulmonary edema. The relative expression levels of inflammation factors were measured by real-time quantitative polymerase chain reaction and Western blotting analysis. The release of neutrophil extracellular traps was observed by immunofluorescence stain. RESULTS In the model group, the gross observation and hematoxylin and eosin staining assay showed the inflammatory cell infiltration, intra-alveolar hemorrhage, and damaged lung tissue structure. The Evans blue dye and bronchoalveolar lavage fluid examination revealed that the lung tissue permeability and edema was significantly increased after injury. Real-time quantitative polymerase chain reaction and Western blotting assays showed that IL-1β, IL-6, TNF-α were upregulated in the model group. Immunofluorescence assay showed that the level of neutrophil extracellular traps in the lung tissue increased significantly in the model group. CONCLUSION The self-made mini shock tube simulation device can be used to establish the animal model of PBLI successfully. Pathological changes of PBLI mice were characterized by mechanical damage and inflammatory response in lung tissue.
Collapse
Affiliation(s)
- Xiang-Yan Meng
- From the Institute of Disaster and Emergency Medicine (X.-Y.M., Q.-Y.L., J.-F.Z., J.-F.L., M.-Y.S., S.-Y.H., S.-F.Y., Y.-M.Z., H.-J.F.), Tianjin University; and Tianjin Key Laboratory of Disaster Medicine Technology (X.-Y.M., Q.-Y.L., J.-F.Z., J.-F.L., M.-Y.S., S.-Y.H., S.-F.Y., Y.-M.Z., H.-J.F.), Tianjin, China
| | - Qian-Ying Lu
- From the Institute of Disaster and Emergency Medicine (X.-Y.M., Q.-Y.L., J.-F.Z., J.-F.L., M.-Y.S., S.-Y.H., S.-F.Y., Y.-M.Z., H.-J.F.), Tianjin University; and Tianjin Key Laboratory of Disaster Medicine Technology (X.-Y.M., Q.-Y.L., J.-F.Z., J.-F.L., M.-Y.S., S.-Y.H., S.-F.Y., Y.-M.Z., H.-J.F.), Tianjin, China
| | - Jian-Feng Zhang
- From the Institute of Disaster and Emergency Medicine (X.-Y.M., Q.-Y.L., J.-F.Z., J.-F.L., M.-Y.S., S.-Y.H., S.-F.Y., Y.-M.Z., H.-J.F.), Tianjin University; and Tianjin Key Laboratory of Disaster Medicine Technology (X.-Y.M., Q.-Y.L., J.-F.Z., J.-F.L., M.-Y.S., S.-Y.H., S.-F.Y., Y.-M.Z., H.-J.F.), Tianjin, China
| | - Jun-Feng Li
- From the Institute of Disaster and Emergency Medicine (X.-Y.M., Q.-Y.L., J.-F.Z., J.-F.L., M.-Y.S., S.-Y.H., S.-F.Y., Y.-M.Z., H.-J.F.), Tianjin University; and Tianjin Key Laboratory of Disaster Medicine Technology (X.-Y.M., Q.-Y.L., J.-F.Z., J.-F.L., M.-Y.S., S.-Y.H., S.-F.Y., Y.-M.Z., H.-J.F.), Tianjin, China
| | - Ming-Yu Shi
- From the Institute of Disaster and Emergency Medicine (X.-Y.M., Q.-Y.L., J.-F.Z., J.-F.L., M.-Y.S., S.-Y.H., S.-F.Y., Y.-M.Z., H.-J.F.), Tianjin University; and Tianjin Key Laboratory of Disaster Medicine Technology (X.-Y.M., Q.-Y.L., J.-F.Z., J.-F.L., M.-Y.S., S.-Y.H., S.-F.Y., Y.-M.Z., H.-J.F.), Tianjin, China
| | - Si-Yu Huang
- From the Institute of Disaster and Emergency Medicine (X.-Y.M., Q.-Y.L., J.-F.Z., J.-F.L., M.-Y.S., S.-Y.H., S.-F.Y., Y.-M.Z., H.-J.F.), Tianjin University; and Tianjin Key Laboratory of Disaster Medicine Technology (X.-Y.M., Q.-Y.L., J.-F.Z., J.-F.L., M.-Y.S., S.-Y.H., S.-F.Y., Y.-M.Z., H.-J.F.), Tianjin, China
| | - Si-Fan Yu
- From the Institute of Disaster and Emergency Medicine (X.-Y.M., Q.-Y.L., J.-F.Z., J.-F.L., M.-Y.S., S.-Y.H., S.-F.Y., Y.-M.Z., H.-J.F.), Tianjin University; and Tianjin Key Laboratory of Disaster Medicine Technology (X.-Y.M., Q.-Y.L., J.-F.Z., J.-F.L., M.-Y.S., S.-Y.H., S.-F.Y., Y.-M.Z., H.-J.F.), Tianjin, China
| | - Yan-Mei Zhao
- From the Institute of Disaster and Emergency Medicine (X.-Y.M., Q.-Y.L., J.-F.Z., J.-F.L., M.-Y.S., S.-Y.H., S.-F.Y., Y.-M.Z., H.-J.F.), Tianjin University; and Tianjin Key Laboratory of Disaster Medicine Technology (X.-Y.M., Q.-Y.L., J.-F.Z., J.-F.L., M.-Y.S., S.-Y.H., S.-F.Y., Y.-M.Z., H.-J.F.), Tianjin, China
| | - Hao-Jun Fan
- From the Institute of Disaster and Emergency Medicine (X.-Y.M., Q.-Y.L., J.-F.Z., J.-F.L., M.-Y.S., S.-Y.H., S.-F.Y., Y.-M.Z., H.-J.F.), Tianjin University; and Tianjin Key Laboratory of Disaster Medicine Technology (X.-Y.M., Q.-Y.L., J.-F.Z., J.-F.L., M.-Y.S., S.-Y.H., S.-F.Y., Y.-M.Z., H.-J.F.), Tianjin, China
| |
Collapse
|
39
|
Shimada S, Shamaa T, Ivanics T, Kitajima T, Collins K, Rizzari M, Yoshida A, Abouljoud M, Moonka D, Lu M, Nagai S. Liver Transplant Recipient Characteristics Associated With Worse Post-Transplant Outcomes in Using Elderly Donors. Transpl Int 2022; 35:10489. [PMID: 36090776 PMCID: PMC9452632 DOI: 10.3389/ti.2022.10489] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 08/10/2022] [Indexed: 12/03/2022]
Abstract
Advanced age of liver donor is a risk factor for graft loss after transplant. We sought to identify recipient characteristics associated with negative post-liver transplant (LT) outcomes in the context of elderly donors. Using 2014–2019 OPTN/UNOS data, LT recipients were classified by donor age: ≥70, 40–69, and <40 years. Recipient risk factors for one-year graft loss were identified and created a risk stratification system and validated it using 2020 OPTN/UNOS data set. At transplant, significant recipient risk factors for one-year graft loss were: previous liver transplant (adjusted hazard ratio [aHR] 4.37, 95%CI 1.98–9.65); mechanical ventilation (aHR 4.28, 95%CI 1.95–9.43); portal thrombus (aHR 1.87, 95%CI 1.26–2.77); serum sodium <125 mEq/L (aHR 2.88, 95%CI 1.34–6.20); and Karnofsky score 10–30% (aHR 2.03, 95%CI 1.13–3.65), 40–60% (aHR 1.65, 95%CI 1.08–2.51). Using those risk factors and multiplying HRs, recipients were divided into low-risk (n = 931) and high-risk (n = 294). Adjusted risk of one-year graft loss in the low-risk recipient group was similar to that of patients with younger donors; results were consistent using validation dataset. Our results show that a system of careful recipient selection can reduce the risks of graft loss associated with older donor age.
Collapse
Affiliation(s)
- Shingo Shimada
- Division of Transplant and Hepatobiliary Surgery, Henry Ford Health System, Detroit, MI, United States
| | - Tayseer Shamaa
- Division of Transplant and Hepatobiliary Surgery, Henry Ford Health System, Detroit, MI, United States
| | - Tommy Ivanics
- Division of Transplant and Hepatobiliary Surgery, Henry Ford Health System, Detroit, MI, United States
| | - Toshihiro Kitajima
- Division of Transplant and Hepatobiliary Surgery, Henry Ford Health System, Detroit, MI, United States
| | - Kelly Collins
- Division of Transplant and Hepatobiliary Surgery, Henry Ford Health System, Detroit, MI, United States
| | - Michael Rizzari
- Division of Transplant and Hepatobiliary Surgery, Henry Ford Health System, Detroit, MI, United States
| | - Atsushi Yoshida
- Division of Transplant and Hepatobiliary Surgery, Henry Ford Health System, Detroit, MI, United States
| | - Marwan Abouljoud
- Division of Transplant and Hepatobiliary Surgery, Henry Ford Health System, Detroit, MI, United States
| | - Dilip Moonka
- Division of Gastroenterology and Hepatology, Henry Ford Health System, Detroit, MI, United States
| | - Mei Lu
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, United States
| | - Shunji Nagai
- Division of Transplant and Hepatobiliary Surgery, Henry Ford Health System, Detroit, MI, United States
- *Correspondence: Shunji Nagai,
| |
Collapse
|
40
|
Li X, Gao Q, Wu W, Hai S, Hu J, You J, Huang D, Wang H, Wu D, Han M, Xi D, Yan W, Chen T, Luo X, Ning Q, Wang X. FGL2-MCOLN3-Autophagy Axis-Triggered Neutrophil Extracellular Traps Exacerbate Liver Injury in Fulminant Viral Hepatitis. Cell Mol Gastroenterol Hepatol 2022; 14:1077-1101. [PMID: 35926777 PMCID: PMC9490102 DOI: 10.1016/j.jcmgh.2022.07.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND & AIMS Fulminant viral hepatitis (FVH) is a life-threatening disease, but its pathogenesis is not fully understood. Neutrophil extracellular traps (NETs) were an unrecognized link between inflammation and coagulation, which are 2 main features of FVH. Here, we investigated the role and mechanism of NETs in the pathogenesis of FVH. METHODS A mouse model of FVH was established by murine hepatitis virus strain-3 infection. Liver leukocytes of infected or uninfected mice were used for single-cell RNA sequencing and whole-transcriptome sequencing. NETs depletion was achieved using DNase 1. Acetaminophen was used to establish a mouse model of non-virus-caused acute liver failure. Clinically, NETs-related markers in liver, plasma, and peripheral neutrophils were assessed in patients with hepatitis B virus (HBV)-related acute liver injury. RESULTS Increased hepatic NETs formation was observed in murine hepatitis virus strain-3-infected mice, but not in acetaminophen-treated mice. NETs depletion improved the liver damage and survival rate in FVH by inhibiting hepatic fibrin deposition and inflammation. An adoptive transfer experiment showed that neutrophil-specific fibrinogen-like protein 2 (FGL2) promoted NETs formation. FGL2 was found to directly interact with mucolipin 3, which regulated calcium influx and initiated autophagy, leading to NETs formation. Clinically, increased plasma NETs level was associated with coagulation dysfunction in patients with HBV acute liver injury. Colocalization of FGL2, NETs, and fibrin in liver was observed in these patients. CONCLUSIONS NETs aggravated liver injury in FVH by promoting fibrin deposition and inflammation. NETs formation was regulated by the FGL2-mucolipin 3-autophagy axis. Targeting NETs may provide a new strategy for the treatment of FVH.
Collapse
Affiliation(s)
- Xitang Li
- Department and Institute of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; National Medical Center for Major Public Health Events, Wuhan, China; State Key Laboratory for Zoonotic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qiang Gao
- Department and Institute of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; National Medical Center for Major Public Health Events, Wuhan, China; State Key Laboratory for Zoonotic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wenhui Wu
- Department and Institute of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; National Medical Center for Major Public Health Events, Wuhan, China; State Key Laboratory for Zoonotic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Suping Hai
- Department and Institute of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; National Medical Center for Major Public Health Events, Wuhan, China; State Key Laboratory for Zoonotic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Junjian Hu
- Department and Institute of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; National Medical Center for Major Public Health Events, Wuhan, China; State Key Laboratory for Zoonotic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jie You
- Department and Institute of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; National Medical Center for Major Public Health Events, Wuhan, China; State Key Laboratory for Zoonotic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Da Huang
- Department and Institute of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; National Medical Center for Major Public Health Events, Wuhan, China; State Key Laboratory for Zoonotic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hongwu Wang
- Department and Institute of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; National Medical Center for Major Public Health Events, Wuhan, China; State Key Laboratory for Zoonotic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Di Wu
- Department and Institute of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; National Medical Center for Major Public Health Events, Wuhan, China; State Key Laboratory for Zoonotic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Meifang Han
- Department and Institute of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; National Medical Center for Major Public Health Events, Wuhan, China; State Key Laboratory for Zoonotic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Dong Xi
- Department and Institute of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; National Medical Center for Major Public Health Events, Wuhan, China; State Key Laboratory for Zoonotic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Weiming Yan
- Department and Institute of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; National Medical Center for Major Public Health Events, Wuhan, China; State Key Laboratory for Zoonotic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tao Chen
- Department and Institute of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; National Medical Center for Major Public Health Events, Wuhan, China; State Key Laboratory for Zoonotic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaoping Luo
- Department and Institute of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qin Ning
- Department and Institute of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; National Medical Center for Major Public Health Events, Wuhan, China; State Key Laboratory for Zoonotic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Xiaojing Wang
- Department and Institute of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; National Medical Center for Major Public Health Events, Wuhan, China; State Key Laboratory for Zoonotic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
41
|
von Meijenfeldt FA, Lisman T. Unravelling the Role of Neutrophil Extracellular Traps in Acute Liver Failure. Cell Mol Gastroenterol Hepatol 2022; 14:720-721. [PMID: 35779579 PMCID: PMC9421577 DOI: 10.1016/j.jcmgh.2022.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 05/25/2022] [Indexed: 12/10/2022]
Affiliation(s)
- Fien A von Meijenfeldt
- Surgical Research Laboratory and Section of Hepatobiliary Surgery and Liver Transplantation Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Ton Lisman
- Surgical Research Laboratory and Section of Hepatobiliary Surgery and Liver Transplantation Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
42
|
Liu L, Ying M, Chen S, Li Q, Chen G, Li H, Mai Z, He Y, Wang B, Xu D, Huang Z, Yan X, Tan N, Chen Z, Liu J, Liu Y. The association between prothrombin time-international normalized ratio and long-term mortality in patients with coronary artery disease: a large cohort retrospective study with 44,662 patients. BMC Cardiovasc Disord 2022; 22:297. [PMID: 35768760 PMCID: PMC9245258 DOI: 10.1186/s12872-022-02619-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 03/24/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The association between prothrombin time-international normalized ratio (PT-INR) and long-term prognosis among patients with coronary artery disease (CAD) without atrial fibrillation or anticoagulant therapy was still unclear. We analyzed the association of PT-INR levels and long-term mortality in a large cohort of CAD patients without atrial fibrillation or using of anticoagulant drugs. METHODS We obtained data from 44,662 patients who were diagnosed with CAD and had follow-up information from January 2008 to December 2018. The patients were divided into 4 groups (Quartile 1: PT-INR ≤ 0.96; Quartile2: 0.96 < PT-INR ≤ 1.01; Quartile3: 1.01 < PT-INR ≤ 1.06; Quartile4: PT-INR > 1.06). The main endpoint was long-term all-cause death. Kaplan-Meier curve analysis and Cox proportional hazards models were used to investigate the association between quartiles of PT-INR levels and long-term all-cause mortality. RESULTS During a median follow-up of 5.25 years, 5613 (12.57%) patients died. We observed a non-linear shaped association between PT-INR levels and long-term all-cause mortality. Patients in high PT-INR level (Quartile4: PT-INR > 1.06) showed a significantly higher long-term mortality than other groups (Quartile2 or 3 or 4), (Compared with Quartile 1, Quartile 2 [0.96 < PT-INR ≤ 1.01], aHR = 1.00, 95% CI 0.91-1.00, P = 0.99; Quartile 3 [1.01 < PT-INR ≤ 1.06], aHR = 1.10, 95% CI 1.01-1.20, P = 0.03; Quartile 4 [PT-INR > 1.06], aHR = 1.33, 95% CI 1.22-1.45, P < 0.05). CONCLUSIONS Our study demonstrates high levels of PT-INR were associated with an increased risk of all-cause mortality.
Collapse
Affiliation(s)
- Liwei Liu
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, South China University of Technology, Guangzhou, 510080, China. .,Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China.
| | - Ming Ying
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, South China University of Technology, Guangzhou, 510080, China
| | - Shiqun Chen
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, South China University of Technology, Guangzhou, 510080, China
| | - Qiang Li
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, South China University of Technology, Guangzhou, 510080, China
| | - Guanzhong Chen
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, South China University of Technology, Guangzhou, 510080, China.,School of Medicine, Guangdong Provincial People's Hospital, South China University of Technology, Guangzhou, 510100, China
| | - Huanqiang Li
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, South China University of Technology, Guangzhou, 510080, China
| | - Ziling Mai
- School of Medicine, Guangdong Provincial People's Hospital, South China University of Technology, Guangzhou, 510100, China
| | - Yibo He
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, South China University of Technology, Guangzhou, 510080, China
| | - Bo Wang
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, South China University of Technology, Guangzhou, 510080, China
| | - Danyuan Xu
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, South China University of Technology, Guangzhou, 510080, China
| | - Zhidong Huang
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, South China University of Technology, Guangzhou, 510080, China
| | - Xiaoming Yan
- Department of Information Technology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Ning Tan
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, South China University of Technology, Guangzhou, 510080, China
| | - Zhujun Chen
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, South China University of Technology, Guangzhou, 510080, China
| | - Jin Liu
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, South China University of Technology, Guangzhou, 510080, China
| | - Yong Liu
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, South China University of Technology, Guangzhou, 510080, China.
| |
Collapse
|
43
|
Ye D, Yao J, Du W, Chen C, Yang Y, Yan K, Li J, Xu Y, Zang S, Zhang Y, Rong X, Zhang R, Xu A, Guo J. Neutrophil Extracellular Traps Mediate Acute Liver Failure in Regulation of miR-223/Neutrophil Elastase Signaling in Mice. Cell Mol Gastroenterol Hepatol 2022; 14:587-607. [PMID: 35660025 PMCID: PMC9307949 DOI: 10.1016/j.jcmgh.2022.05.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 05/20/2022] [Accepted: 05/26/2022] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Marked enhancement of neutrophil infiltration in the liver is a hallmark of acute liver failure (ALF), a severe life-threatening disease with varying etiologies. However, the mechanisms and pathophysiological role corresponding to hepatic neutrophil infiltration during ALF development remain poorly characterized. METHODS Experimental ALF was induced in 10-week-old male microRNA-223 (miR-223) knockout (KO) mice, neutrophil elastase (NE) KO mice, and wild-type controls by intraperitoneal injection of galactosamine hydrochloride and lipopolysaccharide. Age-matched mice were injected with phosphate-buffered saline and served as vehicle controls. RESULTS Mouse liver with ALF showed evident formation of neutrophil extracellular traps (NETs), which were enhanced markedly in miR-223 KO mice. The blockade of NETs by pharmacologic inhibitor GSK484 significantly attenuated neutrophil infiltration and massive necrosis in mouse liver with ALF. ALF-related hepatocellular damage and mortality in miR-223 KO mice were aggravated significantly and accompanied by potentiated neutrophil infiltration in the liver when compared with wild-type controls. Transcriptomic analyses showed that miR-223 deficiency in bone marrow predominantly caused the enrichment of pathways involved in neutrophil degranulation. Likewise, ALF-induced hepatic NE enrichment was potentiated in miR-223 KO mice. Genetic ablation of NE blunted the formation of NETs in parallel with significant attenuation of ALF in mice. Pharmaceutically, pretreatment with the NE inhibitor sivelestat protected mice against ALF. CONCLUSIONS The present study showed the miR-223/NE axis as a key modulator of NETs, thereby exacerbating oxidative stress and neutrophilic inflammation to potentiate hepatocellular damage and liver necrosis in ALF development, and offering potential targets against ALF.
Collapse
Affiliation(s)
- Dewei Ye
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China,Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, China,Guangdong Traditional Chinese Medicine Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China,Key Laboratory of Metabolic Phenotyping in Model Animals, Guangdong Pharmaceutical University, Guangzhou, China,Correspondence Address correspondence to: Dewei Ye, PhD, Lab 406, 4th Floor, Science and Technology Building, Guangdong Pharmaceutical University, 280 Waihuan East Road, Guangzhou Higher Education Mega, Guangzhou, 510006, China.
| | - Jianyu Yao
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China,Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, China,Guangdong Traditional Chinese Medicine Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China,Key Laboratory of Metabolic Phenotyping in Model Animals, Guangdong Pharmaceutical University, Guangzhou, China
| | - Wenfa Du
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China,Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, China,Guangdong Traditional Chinese Medicine Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China,Key Laboratory of Metabolic Phenotyping in Model Animals, Guangdong Pharmaceutical University, Guangzhou, China
| | - Cuishan Chen
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China,Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, China,Guangdong Traditional Chinese Medicine Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China,Key Laboratory of Metabolic Phenotyping in Model Animals, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yong Yang
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China,Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, China,Guangdong Traditional Chinese Medicine Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China,Key Laboratory of Metabolic Phenotyping in Model Animals, Guangdong Pharmaceutical University, Guangzhou, China
| | - Kaixuan Yan
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China,Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, China,Guangdong Traditional Chinese Medicine Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China,Key Laboratory of Metabolic Phenotyping in Model Animals, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jufei Li
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China,Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, China,Guangdong Traditional Chinese Medicine Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China,Key Laboratory of Metabolic Phenotyping in Model Animals, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ying Xu
- School of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Shufei Zang
- Department of Endocrinology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Yuying Zhang
- Central Laboratory, Shenzhen Longhua Maternity and Child Healthcare Hospital, Shenzhen, China
| | - Xianglu Rong
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China,Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, China,Guangdong Traditional Chinese Medicine Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China
| | - Rongxin Zhang
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Institute of Basic Medical Sciences, Department of Biotechnology, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong, China,Prof. Aimin Xu, State Key Laboratory of Pharmaceutical Biotechnology, and Department of Medicine, The University of Hong Kong, Room L8-39, Lab Block, 21 Sassoon Road, Hong Kong. Fax: +00852-2816 2095.
| | - Jiao Guo
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China,Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, China,Guangdong Traditional Chinese Medicine Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China,Prof. Jiao Guo, Room 403, 4th Floor, Science and Technology Building, Guangdong Pharmaceutical University, 280 Waihuan East Road, Guangzhou Higher Education Mega, 510006, China.
| |
Collapse
|
44
|
Liu G, Fan X, Yang L, Luo X. Letter to the editor: Etiology and coagulation should be considered more. Hepatology 2022; 75:769-770. [PMID: 34923654 DOI: 10.1002/hep.32287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 11/18/2021] [Indexed: 02/05/2023]
Affiliation(s)
- Guofeng Liu
- Department of Gastroenterology and Hepatology, Sichuan University-University of Oxford Huaxi Joint Center for Gastrointestinal Cancer, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoli Fan
- Department of Gastroenterology and Hepatology, Sichuan University-University of Oxford Huaxi Joint Center for Gastrointestinal Cancer, West China Hospital, Sichuan University, Chengdu, China
| | - Li Yang
- Department of Gastroenterology and Hepatology, Sichuan University-University of Oxford Huaxi Joint Center for Gastrointestinal Cancer, West China Hospital, Sichuan University, Chengdu, China
| | - Xuefeng Luo
- Department of Gastroenterology and Hepatology, Sichuan University-University of Oxford Huaxi Joint Center for Gastrointestinal Cancer, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
45
|
Hilscher MB, Shah VH. A NET gain in our understanding of acute liver failure. Hepatology 2022; 75:511-513. [PMID: 34890053 DOI: 10.1002/hep.32278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 12/06/2021] [Indexed: 12/08/2022]
Affiliation(s)
- Moira B Hilscher
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Vijay H Shah
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|