1
|
Wu L, Li J, Zou J, Tang D, Chen R. Vagus nerve modulates acute-on-chronic liver failure progression via CXCL9. Chin Med J (Engl) 2025; 138:1103-1115. [PMID: 38945689 PMCID: PMC12068771 DOI: 10.1097/cm9.0000000000003104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Indexed: 07/02/2024] Open
Abstract
BACKGROUND Hepatic inflammatory cell accumulation and the subsequent systematic inflammation drive acute-on-chronic liver failure (ACLF) development. Previous studies showed that the vagus nerve exerts anti-inflammatory activity in many inflammatory diseases. Here, we aimed to identify the key molecule mediating the inflammatory process in ACLF and reveal the neuroimmune communication arising from the vagus nerve and immunological disorders of ACLF. METHODS Proteomic analysis was performed and validated in ACLF model mice or patients, and intervention animal experiments were conducted using neutralizing antibodies. PNU-282987 (acetylcholine receptor agonist) and vagotomy were applied for perturbing vagus nerve activity. Single-cell RNA sequencing (scRNA-seq), flow cytometry, immunohistochemical and immunofluorescence staining, and clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9 (CRISPR/Cas9) technology were used for in vivo or in vitro mechanistic studies. RESULTS The unbiased proteomics identified C-X-C motif chemokine ligand 9 (CXCL9) as the greatest differential protein in the livers of mice with ACLF and its relation to the systematic inflammation and mortality were confirmed in patients with ACLF. Interventions on CXCL9 and its receptor C-X-C chemokine receptor 3 (CXCR3) improved liver injury and decreased mortality of ACLF mice, which were related to the suppressing of hepatic immune cells' accumulation and activation. Vagus nerve stimulation attenuated while vagotomy aggravated the expression of CXCL9 and the severity of ACLF. Blocking CXCL9 and CXCR3 ameliorated liver inflammation and increased ACLF-associated mortality in ACLF mice with vagotomy. scRNA-seq revealed that hepatic macrophages served as the major source of CXCL9 in ACLF and were validated by immunofluorescence staining and flow cytometry analysis. Notably, the expression of CXCL9 in macrophages was modulated by vagus nerve-mediated cholinergic signaling. CONCLUSIONS Our novel findings highlighted that the neuroimmune communication of the vagus nerve-macrophage-CXCL9 axis contributed to ACLF development. These results provided evidence for neuromodulation as a promising approach for preventing and treating ACLF.
Collapse
Affiliation(s)
- Li Wu
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jie Li
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Ju Zou
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Ruochan Chen
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
2
|
Jia L, He WP, Xing HC, Li J, Yu HW, Hou W, Xue R, Zhao J, Meng QH. The restoration of immunity characterized by the recovery of myeloid dendritic cells represent a favorable response to methylprednisolone therapy for HBV-ACLF patients: A prospective cohort study. Cytokine 2025; 189:156894. [PMID: 40043628 DOI: 10.1016/j.cyto.2025.156894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/04/2024] [Accepted: 02/12/2025] [Indexed: 03/18/2025]
Abstract
BACKGROUND The use of methylprednisolone (MP) is still controversial for hepatitis B virus related acute-on-chronic liver failure (HBV-ACLF). We aimed to explore the change in dendritic cells (DCs) during MP treatment in HBV-ACLF to guide the use of MP to improve patient's prognosis. METHODS Patients with HBV-ACLF were prospectively allocated to groups given methylprednisolone intravenously (1.5 mg/kg/day for the first 3 days, 1 mg/kg/day for the second 2 days, and 0.5 mg/kg/day for the last 2 days, MP group, n = 36) plus standard treatment or standard treatment only (CM group, n = 34). The phenotype [myeloid and plasmacytoid DCs (mDCs, pDCs)] and function of DCs (IL-12 and IFN-α production) were measured at baseline (0d), 3d, 7d, 10d, 14d, 28d, and then monthly until 3 months. Patients' survival was assessed until day 90. RESULTS The 3-month survival rate was significantly higher in the MP group than the control (72.0 % vs. 35.5 %,P < 0.01). The phenotype and function of DCs were suppressed in the MP group. The mDCs counts was lower in non-survivors compared to survivors at baseline. Patients with a decline in mDCs counts at the 7th day and a continuous increase in mDCs counts from the 10th day presented a better outcome for patients with MP treatment. Bilirubin was the only relative factor for the restoration of mDCs in the MP group (odds ratio, 0.991; 95 % confidence interval, 0.984-0.999; P = 0.023). CONCLUSIONS Methylprednisolone could improve the outcome of HBV-ACLF by inhibiting the circulating mDCs counts. And the recovery of mDCs counts after methylprednisolone treatment could represent a favorable response. We can consider monitoring the circulating DCs counts to guide the use of MP in HBV-ACLF in order to improve patient outcomes.
Collapse
Affiliation(s)
- Lin Jia
- Department of Infection and Immunity, Beijing You-An Hospital, Capital Medical University, Beijing, China
| | - Wei-Ping He
- 302 Hospital of People's Liberation Army, Liver Disease Center for Military Staff, Beijing, China
| | - Hui-Chun Xing
- Institute of Infectious Diseases, Beijing DiTan Hospital, Capital Medical University, Beijing, China
| | - Juan Li
- Department of Medical Oncology, Beijing You-An Hospital, Capital Medical University, Beijing, China
| | - Hong-Wei Yu
- Outpatient Department, Beijing You-An Hospital, Capital Medical University, Beijing, China
| | - Wei Hou
- Department I, Center for Liver Diseases, Beijing You-An Hospital, Capital Medical University, Beijing, China
| | - Ran Xue
- Key laboratory of Carcinogenesis and Translational Research, Department of phase I clinical trial, Peking University Cancer Hospital & Institute, Beijing, China
| | - Juan Zhao
- Department II, Center for Liver Diseases, Beijing You-An Hospital, Capital Medical University, Beijing, China
| | - Qing-Hua Meng
- Department of Medical Oncology, Beijing You-An Hospital, No. 8 You An Men Wai Street, Fengtai District, Beijing 100069, China.
| |
Collapse
|
3
|
Li T, Adams J, Zhu P, Zhang T, Tu F, Gravitte A, Zhang X, Liu L, Casteel J, Yakubenko V, Williams DL, Li C, Wang X. The role of heme in sepsis induced Kupffer cell PANoptosis and senescence. Cell Death Dis 2025; 16:284. [PMID: 40221420 PMCID: PMC11993645 DOI: 10.1038/s41419-025-07637-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 03/18/2025] [Accepted: 04/04/2025] [Indexed: 04/14/2025]
Abstract
Elevated heme levels, a consequence of hemolysis, are strongly associated with increased susceptibility to bacterial infections and adverse sepsis outcomes, particularly in older populations. However, the underlying mechanisms remain poorly understood. Using a cecal ligation and puncture (CLP) model of sepsis, we demonstrate that elevated heme levels correlate with Kupffer cell loss, increased bacterial burden, and heightened mortality. Mechanistically, we identify mitochondrial damage as a key driver of heme- and bacterial-induced Kupffer cell PANoptosis, a form of cell death integrating pyroptosis, apoptosis, and necroptosis, as well as cellular senescence. Specifically, heme activates phospholipase C gamma (PLC-γ), facilitating the translocation of cleaved gasdermin D (c-GSDMD) to mitochondria, resulting in GSDMD pore formation, mitochondrial dysfunction, and the release of mitochondrial DNA (mtDNA) during bacterial infection. This mitochondrial damage amplifies PANoptosis and triggers the cGAS-STING signaling pathway, further driving immune senescence. Notably, PLC-γ inhibition significantly reduces mitochondrial damage, cell death, and senescence caused by heme and bacterial infection. Furthermore, we show that hemopexin, a heme scavenger, effectively mitigates sepsis-induced Kupffer cell death and senescence, enhances bacterial clearance, and improves survival outcomes in both young and aged mice. These findings establish mitochondrial damage as a central mediator of heme induced Kupffer cell loss and highlight PLC-γ inhibition and hemopexin administration as promising therapeutic strategies for combating sepsis associated immune dysfunction.
Collapse
Affiliation(s)
- Tingting Li
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Joseph Adams
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Peilin Zhu
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Tao Zhang
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Fei Tu
- UMPC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15232, USA
| | - Amy Gravitte
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Xiaojin Zhang
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Li Liu
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jared Casteel
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Valentin Yakubenko
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
- Center of Excellence in Inflammation, Infectious Disease, and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - David L Williams
- Center of Excellence in Inflammation, Infectious Disease, and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
- Department of Surgery, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Chuanfu Li
- Center of Excellence in Inflammation, Infectious Disease, and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
- Department of Surgery, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Xiaohui Wang
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA.
- Center of Excellence in Inflammation, Infectious Disease, and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA.
| |
Collapse
|
4
|
Baquero C, Iniesta‐González M, Palao N, Fernández‐Infante C, Cueto‐Remacha M, Mancebo J, de la Cámara‐Fuentes S, Rodrigo‐Faus M, Valdecantos MP, Valverde AM, Sequera C, Manzano S, Cuesta ÁM, Gutierrez‐Uzquiza A, Bragado P, Guerrero C, Porras A. Platelet C3G protects from liver fibrosis, while enhancing tumor growth through regulation of the immune response. J Pathol 2025; 265:502-517. [PMID: 39989399 PMCID: PMC11880977 DOI: 10.1002/path.6403] [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: 10/28/2024] [Revised: 11/29/2024] [Accepted: 01/09/2025] [Indexed: 02/25/2025]
Abstract
Primary liver cancer usually occurs in the context of chronic liver disease (CLD), in association with fibrosis. Platelets have emerged as important regulators of CLD and liver cancer, although their precise function and mechanism of action need to be clarified. C3G (RapGEF1) regulates platelet activation, adhesion, and secretion. Here we evaluate the role of platelet C3G in chemically induced fibrosis and liver cancer associated with fibrosis using genetically modified mouse models. We found that while overexpression of full-length C3G in platelets decreased liver fibrosis induced by chronic treatment with CCl4, overexpressed C3G lacking the catalytic domain did not, although in both cases platelet recruitment to the liver was similar. In addition, C3G deletion in platelets (PF4-C3GKO mouse model) increased CCl4-induced liver damage and hepatic fibrosis, reducing liver platelets and macrophages. Moreover, early liver immune response to CCl4 was altered in PF4-C3GKO mice, with a remarkable lower activation of macrophages and increased monocyte-derived macrophages compared to WT mice. On the other hand, in response to DEN+CCl4, PF4-C3G WT mice exhibited more and larger liver tumors than PF4-C3GKO mice, accompanied by the presence of more platelets, despite having less fibrosis in previous steps. Liver immune cell populations were also differentially regulated in PF4-C3GKO mice, highlighting the higher number of macrophages, likely with a pro-inflammatory phenotype, present in the liver in response to chronic DEN+CCl4 treatment. Proteins upregulated or downregulated in platelet-rich plasma from PF4-C3GKO compared to WT mice might regulate the immune response and tumor development. In this regard, enrichment analyses using proteomic data showed changes in several proteins involved in platelet activation and immune response pathways. Additionally, the higher secretion of CD40L by PF4-C3GKO platelets could contribute to their antitumor effect. Therefore, platelet C3G presents antifibrotic and protumor effects in the liver, likely mediated by changes in the immune response. © 2025 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Cristina Baquero
- Departamento de Bioquímica y Biología Molecular, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC)MadridSpain
| | - Minerva Iniesta‐González
- Departamento de Bioquímica y Biología Molecular, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC)MadridSpain
| | - Nerea Palao
- Departamento de Bioquímica y Biología Molecular, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC)MadridSpain
| | - Cristina Fernández‐Infante
- Instituto de Biología Molecular y Celular del Cáncer (IMBCC)Universidad de Salamanca‐CSICSalamancaSpain
- Instituto de Investigación Biomédica de Salamanca (IBSAL)SalamancaSpain
| | - Mateo Cueto‐Remacha
- Departamento de Bioquímica y Biología Molecular, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC)MadridSpain
| | - Jaime Mancebo
- Departamento de Bioquímica y Biología Molecular, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC)MadridSpain
| | | | - María Rodrigo‐Faus
- Departamento de Bioquímica y Biología Molecular, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC)MadridSpain
| | - M Pilar Valdecantos
- Instituto de Investigaciones Biomédicas (IIBM) Alberto Sols‐Morreale (CSIC‐UAM)MadridSpain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem)Instituto de Salud Carlos IIIMadridSpain
| | - Angela M Valverde
- Instituto de Investigaciones Biomédicas (IIBM) Alberto Sols‐Morreale (CSIC‐UAM)MadridSpain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem)Instituto de Salud Carlos IIIMadridSpain
| | - Celia Sequera
- Departamento de Bioquímica y Biología Molecular, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
- Aix Marseille Univ, CNRS, InsermInstitut Paoli‐Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM)MarseilleFrance
| | - Sara Manzano
- Departamento de Bioquímica y Biología Molecular, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
| | - Ángel M Cuesta
- Departamento de Bioquímica y Biología Molecular, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC)MadridSpain
| | - Alvaro Gutierrez‐Uzquiza
- Departamento de Bioquímica y Biología Molecular, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC)MadridSpain
| | - Paloma Bragado
- Departamento de Bioquímica y Biología Molecular, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC)MadridSpain
| | - Carmen Guerrero
- Instituto de Biología Molecular y Celular del Cáncer (IMBCC)Universidad de Salamanca‐CSICSalamancaSpain
- Instituto de Investigación Biomédica de Salamanca (IBSAL)SalamancaSpain
- Departamento de MedicinaUniversidad de SalamancaSalamancaSpain
| | - Almudena Porras
- Departamento de Bioquímica y Biología Molecular, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC)MadridSpain
| |
Collapse
|
5
|
Fernandez Alarcon J, Perez Schmidt P, Panini N, Caruso F, Violatto MB, Sukubo NG, Martinez‐Serra A, Ekalle‐Soppo CB, Morelli A, Moscatiello GY, Grasselli C, Corbelli A, Fiordaliso F, Kelk J, Petrosilli L, d'Orazio G, Mateu Ferrando R, Verdaguer Ferrer A, Fornaguera C, Lay L, Fumagalli S, Recchia S, Monopoli MP, Polito L, Bigini P, Sitia G. Functional Polarization of Liver Macrophages by Glyco Gold Nanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407458. [PMID: 39950558 PMCID: PMC12021048 DOI: 10.1002/advs.202407458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/24/2024] [Indexed: 04/26/2025]
Abstract
Macrophages are crucial drivers of innate immunity. Reprogramming macrophages to a restorative phenotype in cancer or autoimmune diseases can stop their cancer-promoting activity or trigger anti-inflammatory immunity. Glycans have emerged as key components for immunity as they are involved in many pathophysiological disorders. Previous studies have demonstrated that supraphysiological amounts of mannose (Man) or sialic acid (Sia) can inhibit tumor growth and stimulate differentiation of regulatory T cells. Man is known to affect glucose metabolism in glycolysis by competing for the same intracellular transporters and affecting macrophage polarization, whereas Sia alters macrophage differentiation via signaling through Siglec-1. Herein, this work describes a macrophage targeting platform using gold nanoparticles (GNPs) functionalized with Man and Sia monosaccharides which exhibit high liver tropism. A single dose of glyco-GNPs can convert macrophages to a restorative phenotype in two completely different immune environments. Man promotes tumor-associated macrophages toward an antitumorigenic activity in a MC38 liver colorectal cancer model by secretion of TNF-α, IL -1β, and IL -6 in the tumor microenvironment. However, in a proinflammatory environment, as observed in a mouse model of autoimmune disease, primary biliary cholangitis, Man impairs the production of TNF-α, IL-1β, Arg1, and IL-6 cytokines. The results probe the dual role of Man in macrophage repolarization in response to the immune system. This study is a proof-of-concept that demonstrates that nanomedicine using specific glycans designed to target other immune cells such as myeloid cells, are a promising strategy not only against cancer but also against other pathologies such as autoimmune diseases.
Collapse
Affiliation(s)
- Jennifer Fernandez Alarcon
- Department of Molecular Biochemistry and PharmacologyIstituto di Ricerche Farmacologiche Mario Negri IRCCSVia Mario Negri 2Milano20156Italy
- Grup d'Enginyeria de Materials (GEMAT)Institut Químic de Sarrià (IQS)Universitat Ramon Llull (URL)Via Augusta 390Barcelona08017Spain
| | - Patricia Perez Schmidt
- Istituto di Scienze e Tecnologie Chimiche “Giulio Natta”SCITEC‐CNR, Via G. Fantoli 16/15Milano20138Italy
| | - Nicolo Panini
- Department of OncologyIstituto di Ricerche Farmacologiche Mario Negri IRCCSVia Mario Negri 2Milano20156Italy
| | - Francesca Caruso
- Experimental Hepatology UnitDivision of Immunology, Transplantation and Infectious DiseasesIRCCS San Raffaele Scientific InstituteVia Olgettina 58Milano20132Italy
| | - Martina B. Violatto
- Department of Molecular Biochemistry and PharmacologyIstituto di Ricerche Farmacologiche Mario Negri IRCCSVia Mario Negri 2Milano20156Italy
| | - Naths Grazia Sukubo
- School of Medicine and SurgeryUniversity of Milano‐BicoccaPiazza dell'Ateneo Nuovo1Milano20126Italy
| | - Alberto Martinez‐Serra
- Department of ChemistryRoyal College of Surgeons of Ireland RCSISt Stephens Green 123DublinIreland
| | - Charlotte Blanche Ekalle‐Soppo
- Experimental Hepatology UnitDivision of Immunology, Transplantation and Infectious DiseasesIRCCS San Raffaele Scientific InstituteVia Olgettina 58Milano20132Italy
| | - Annalisa Morelli
- Department of Molecular Biochemistry and PharmacologyIstituto di Ricerche Farmacologiche Mario Negri IRCCSVia Mario Negri 2Milano20156Italy
| | - Giulia Yuri Moscatiello
- Department of Molecular Biochemistry and PharmacologyIstituto di Ricerche Farmacologiche Mario Negri IRCCSVia Mario Negri 2Milano20156Italy
| | - Chiara Grasselli
- Department of OncologyIstituto di Ricerche Farmacologiche Mario Negri IRCCSVia Mario Negri 2Milano20156Italy
| | - Alessandro Corbelli
- Department of Molecular Biochemistry and PharmacologyIstituto di Ricerche Farmacologiche Mario Negri IRCCSVia Mario Negri 2Milano20156Italy
| | - Fabio Fiordaliso
- Department of Molecular Biochemistry and PharmacologyIstituto di Ricerche Farmacologiche Mario Negri IRCCSVia Mario Negri 2Milano20156Italy
| | - Joe Kelk
- Department of NeurosciencesIstituto di Ricerche Farmacologiche Mario Negri IRCCSVia Mario Negri 2Milano20156Italy
| | - Laura Petrosilli
- Department of Organic ChemistryUniversity degli Studi di MilanoVia Golgi 19Milano20133Italy
| | - Giuseppe d'Orazio
- Department of Organic ChemistryUniversity degli Studi di MilanoVia Golgi 19Milano20133Italy
| | - Ruth Mateu Ferrando
- Department of Organic ChemistryUniversity degli Studi di MilanoVia Golgi 19Milano20133Italy
| | - Ariadna Verdaguer Ferrer
- Department of Analytical and Applied ChemistryInstitut Químic de Sarrià (IQS)Universitat Ramon Llull (URL)Via Augusta 390Barcelona08017Spain
| | - Cristina Fornaguera
- Grup d'Enginyeria de Materials (GEMAT)Institut Químic de Sarrià (IQS)Universitat Ramon Llull (URL)Via Augusta 390Barcelona08017Spain
| | - Luigi Lay
- Department of Organic ChemistryUniversity degli Studi di MilanoVia Golgi 19Milano20133Italy
| | - Stefano Fumagalli
- Department of NeurosciencesIstituto di Ricerche Farmacologiche Mario Negri IRCCSVia Mario Negri 2Milano20156Italy
| | - Sandro Recchia
- Department of Science and High TechnologyUniversity of InsubriaVia Valleggio 11Como22100Italy
| | - Marco P. Monopoli
- Department of ChemistryRoyal College of Surgeons of Ireland RCSISt Stephens Green 123DublinIreland
| | - Laura Polito
- Istituto di Scienze e Tecnologie Chimiche “Giulio Natta”SCITEC‐CNR, Via G. Fantoli 16/15Milano20138Italy
| | - Paolo Bigini
- Department of Molecular Biochemistry and PharmacologyIstituto di Ricerche Farmacologiche Mario Negri IRCCSVia Mario Negri 2Milano20156Italy
| | - Giovanni Sitia
- Experimental Hepatology UnitDivision of Immunology, Transplantation and Infectious DiseasesIRCCS San Raffaele Scientific InstituteVia Olgettina 58Milano20132Italy
| |
Collapse
|
6
|
Wang Z, Zhong D, Yan T, Zheng Q, Zhou E, Ye Z, He X, Liu Y, Yan J, Yuan Y, Wang Y, Cai X. Stem Cells from Human Exfoliated Deciduous Teeth-Derived Exosomes for the Treatment of Acute Liver Injury and Liver Fibrosis. ACS APPLIED MATERIALS & INTERFACES 2025; 17:17948-17964. [PMID: 40087139 PMCID: PMC11955941 DOI: 10.1021/acsami.4c19748] [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] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 03/06/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
Mesenchymal stem cells (MSCs) play a crucial role in regenerative medicine due to their regenerative potential. However, traditional MSC-based therapies are hindered by issues such as microvascular obstruction and low cell survival after transplantation. Exosomes derived from MSCs (MSC-Exo) provide a cell-free, nanoscale alternative, mitigating these risks and offering therapeutic potential for liver diseases. Nonetheless, the functional variability of MSCs from different sources complicates their clinical application. Stem cells derived from human exfoliated deciduous teeth (SHED) offer advantages such as ease of procurement and robust proliferative capacity, but their secretome, particularly SHED-Exo, remains underexplored in the context of liver disease therapy. This study analyzed MSC-Exo from various sources via small RNA sequencing to identify differences in microRNA profiles, aiding in the selection of optimal MSC sources for clinical use. SHED-Exo was subsequently tested in an acute liver injury model, showing notable regenerative effects, including enhanced hepatocyte proliferation, macrophage polarization, and reduced inflammation. Despite strong liver-targeting properties, the rapid hepatic clearance of SHED-Exo limits its effectiveness in chronic liver diseases. To address this challenge, a GelMA-based hydrogel was developed for in situ delivery, ensuring sustained release and enhanced antifibrotic efficacy, providing a promising strategy for chronic liver disease management.
Collapse
Affiliation(s)
- Ziyuan Wang
- Department
of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Danyang Zhong
- Department
of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Tingting Yan
- Department
of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Qiang Zheng
- Department
of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Enjie Zhou
- Department
of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Zhichao Ye
- Department
of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Xiaoyan He
- Department
of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Yu Liu
- Department
of Cardiac Surgery, Zhejiang University
School of Medicine Sir Run Run Shaw Hospital, Hangzhou 310016, Zhejiang, China
| | - Jianing Yan
- Department
of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Yuyang Yuan
- Department
of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Yifan Wang
- Department
of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
- National
Engineering Research Center of Innovation and Application of Minimally
Invasive Instruments, Hangzhou 310016, China
- Zhejiang
Minimal Invasive Diagnosis and Treatment Technology Research Center
of Severe Hepatobiliary Disease, Hangzhou 310016, China
| | - Xiujun Cai
- Department
of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
- National
Engineering Research Center of Innovation and Application of Minimally
Invasive Instruments, Hangzhou 310016, China
- Zhejiang
Minimal Invasive Diagnosis and Treatment Technology Research Center
of Severe Hepatobiliary Disease, Hangzhou 310016, China
| |
Collapse
|
7
|
Larsen FS, Saliba F. Liver support systems and liver transplantation in acute liver failure. Liver Int 2025; 45:e15633. [PMID: 37288706 PMCID: PMC11815598 DOI: 10.1111/liv.15633] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/15/2023] [Accepted: 05/24/2023] [Indexed: 06/09/2023]
Abstract
Acute liver failure (ALF) results in a multitude of complications that result in multi-organ failure. This review focuses on the pathophysiological processes and how to manage with these with artificial liver support and liver transplantation (LT). The pathophysiological sequence of events behind clinical deterioration in ALF comes down to two profound consequences of the failing liver. The first is the development of hyperammonemia, as the liver can no longer synthesize urea. The result is that the splanchnic system instead of removing ammonia becomes an ammonia-producing organ system that causes hepatic encephalopathy (HE) and cerebral oedema. The second complication is caused by the necrotic liver cells that release large molecules that originate from degrading proteins, that is damage associated molecular patterns (DAMPs) which causes inflammatory activation of intrahepatic macrophages and an overflow of DAMPs molecules into the systemic circulation resulting in a clinical picture that resembles septic shock. In this context the combined use of continuous renal replacement therapy (CRRT) and plasma exchange are rational and simple ways to remove ammonia and DAMPS molecules. This combination improve survival for ALF patients deemed not appropriate for LT, despite poor prognostic criteria, but also ensure a better stability of vital organs while awaiting LT. The combination of CRRT with albumin dialysis tends to have a similar effect. Currently, the selection criteria for LT for non-paracetamol cases appear robust while the criteria for paracetamol-intoxicated patients have become more unreliable and now consist of more dynamic prognostic systems. For patients that need LT for survival, a tremendous improvement in the post-LT results has been achieved during the last decade with a survival that now reach merely 90% which is mirroring the results seen after LT for chronic liver disease.
Collapse
Affiliation(s)
- Fin S. Larsen
- Department of Intestinal Failure and Liver DiseasesRigshospitalet, University Hospital CopenhagenCopenhagenDenmark
| | - Faouzi Saliba
- AP‐HP Hôpital Paul Brousse, Hepato‐Biliary Center and Liver Transplant ICUUniversity Paris Saclay, INSERM unit N°1193VillejuifFrance
| |
Collapse
|
8
|
Morrison MA, Artru F, Trovato FM, Triantafyllou E, McPhail MJ. Potential therapies for acute-on-chronic liver failure. Liver Int 2025; 45:e15545. [PMID: 36800487 PMCID: PMC11815631 DOI: 10.1111/liv.15545] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/16/2023] [Accepted: 02/15/2023] [Indexed: 02/19/2023]
Abstract
Acute-on-chronic liver failure (ACLF) is a syndrome that develops in approximately 30% of patients hospitalised with cirrhosis and is characterised by an acute decompensation of liver function associated with extra-hepatic organ failures and a high short-term mortality. At present, no specific therapies are available for ACLF, and current management is limited to treatment of the precipitating event and organ support. Given the high prevalence and high mortality of this severe liver disease, there is an urgent need for targeted treatments. There is increasing evidence of the important role played by systemic inflammation and immune dysfunction in the pathophysiology of ACLF and a better understanding of these immune processes is resulting in new therapeutic targets. The aim of this review is to present an overview of ongoing studies of potentially promising therapies and how they could be utilised in the management of ACLF.
Collapse
Affiliation(s)
- Maura A. Morrison
- Institute of Liver StudiesKing's College HospitalLondonUK
- Department of Inflammation Biology, School of Immunology and Microbial SciencesKing's College LondonLondonUK
| | - Florent Artru
- Institute of Liver StudiesKing's College HospitalLondonUK
- Department of Inflammation Biology, School of Immunology and Microbial SciencesKing's College LondonLondonUK
| | - Francesca M. Trovato
- Institute of Liver StudiesKing's College HospitalLondonUK
- Department of Inflammation Biology, School of Immunology and Microbial SciencesKing's College LondonLondonUK
| | - Evangelos Triantafyllou
- Section of Hepatology and Gastroenterology, Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
| | - Mark J. McPhail
- Institute of Liver StudiesKing's College HospitalLondonUK
- Department of Inflammation Biology, School of Immunology and Microbial SciencesKing's College LondonLondonUK
| |
Collapse
|
9
|
Damasceno ROS, Pinheiro JLS, da Silva LD, Rodrigues LHM, Emídio JJ, Lima TC, de Sousa DP. Phytochemistry and Anti-Inflammatory and Antioxidant Activities of Cinnamomum osmophloeum and Its Bioactive Constituents: A Review. PLANTS (BASEL, SWITZERLAND) 2025; 14:562. [PMID: 40006821 PMCID: PMC11859615 DOI: 10.3390/plants14040562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/31/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025]
Abstract
Cinnamomum osmophloeum, commonly known as indigenous cinnamon, is a tree species native to Taiwan's hardwood forests. It has been extensively investigated for its chemical composition and bioactivities. Several reports have shown that C. osmophloeum leaves are rich in aromatic oils, which are grouped into various chemotypes based on their major constituents. Components of the volatile oils included phenylpropanoids, monoterpenoids, sesquiterpenoids, phenols, coumarins, and other miscellaneous compounds. In addition, other secondary metabolites previously identified in this species included flavonol glycosides, phenolic acids, lignans, proanthocyanidins, and cyclopropanoids. C. osmophloeum is widely recognized for its medicinal and industrial applications, particularly its essential oils. In general, essential oils exhibit remarkable anti-inflammatory and antioxidant actions, enabling them to modulate key inflammatory mediators and neutralize free radicals. This review explored the phytochemical composition of the essential oils and extracts from C. osmophloeum as well as therapeutic potential of this species, focusing on the action mechanisms and clinical potential. We hope that this review will contribute to a better understanding of the biological effects of this plant and its potential applications in the management of conditions associated with inflammation and oxidative stress.
Collapse
Affiliation(s)
- Renan Oliveira Silva Damasceno
- Department of Physiology and Pharmacology, Federal University of Pernambuco, Recife 50670-901, Pernambuco, Brazil; (J.L.S.P.); (L.D.d.S.); (L.H.M.R.)
| | - João Lucas Silva Pinheiro
- Department of Physiology and Pharmacology, Federal University of Pernambuco, Recife 50670-901, Pernambuco, Brazil; (J.L.S.P.); (L.D.d.S.); (L.H.M.R.)
| | - Lorena Duarte da Silva
- Department of Physiology and Pharmacology, Federal University of Pernambuco, Recife 50670-901, Pernambuco, Brazil; (J.L.S.P.); (L.D.d.S.); (L.H.M.R.)
| | - Lucas Henrique Marques Rodrigues
- Department of Physiology and Pharmacology, Federal University of Pernambuco, Recife 50670-901, Pernambuco, Brazil; (J.L.S.P.); (L.D.d.S.); (L.H.M.R.)
| | - Jeremias Justo Emídio
- Department of Pharmaceutical Sciences, Federal University of Paraíba, João Pessoa 58051-970, Paraíba, Brazil;
| | - Tamires Cardoso Lima
- Department of Pharmacy, Federal University of Sergipe, São Cristóvão 49100-000, Sergipe, Brazil;
| | - Damião Pergentino de Sousa
- Department of Pharmaceutical Sciences, Federal University of Paraíba, João Pessoa 58051-970, Paraíba, Brazil;
| |
Collapse
|
10
|
Xiong S, Xie J, Xiang F, Yu J, Li Y, Xia B, Zhang Z, Li C, Lin L. Research progress on pharmacological effects against liver and eye diseases of flavonoids present in Chrysanthum indicum L., Chrysanthemum morifolium Ramat., Buddleja officinalis Maxim. and Sophora japonica L. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119094. [PMID: 39532220 DOI: 10.1016/j.jep.2024.119094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/07/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Chrysanthemum indicum L., Chrysanthemum morifolium Ramat., Buddleja officinalis Maxim., and Sophora japonica L. have the effects of "Clearing the liver" and "Improving vision". Flavonoids are their main active ingredients, but there are few reports on their simultaneous liver and eye protective effects. AIM OF THE STUDY Overview of the role of flavonoids of the four medicinal flowers (FFMF) in the prevention and treatment of liver and eye diseases. MATERIALS AND METHODS The Web of Science, PubMed, CNKI, Google Scholar, and WanFang databases were searched for FFMF. Using "hepatitis", "liver fibrosis", "liver cancer", "dry eye syndrome", "cataracts", "glaucoma", "age-related macular degeneration", and "diabetic retinopathy" as the keywords, we summarized the main pathological mechanisms of these diseases and the role of FFMF in their prevention and treatment. RESULTS We found that the four medicinal flowers contained a total of 125 flavonoids. They can maintain liver and eye homeostasis by regulating pathological mechanisms such as oxidative stress, inflammation, endoplasmic reticulum stress, mitochondrial dysfunction, glucose and lipid metabolism disorders, and programmed cell death, exerting the effect of "clearing the liver and improving vision". CONCLUSION FFMF have a series of beneficial properties such as antioxidant, anti-inflammatory, antiviral, and antifibrotic activity, and the regulation of angiogenesis, glycolipid metabolism and programmed cell death, which may explain the efficacy of the four traditional Chinese medicines for "Clearing the liver" and "Improving vision".
Collapse
Affiliation(s)
- Suhui Xiong
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China; Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, China.
| | - Jingchen Xie
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China; Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, China.
| | - Feng Xiang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China; Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, China.
| | - Jiahui Yu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China; Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, China.
| | - Yamei Li
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China; Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, China.
| | - Bohou Xia
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China; Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, China.
| | - Zhimin Zhang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China; Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, China.
| | - Chun Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Limei Lin
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China; Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, China.
| |
Collapse
|
11
|
Wu X, Pan T, Fang Z, Hui T, Yu X, Liu C, Guo Z, Liu C. Identification of EGR1 as a Key Diagnostic Biomarker in Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) Through Machine Learning and Immune Analysis. J Inflamm Res 2025; 18:1639-1656. [PMID: 39925925 PMCID: PMC11806694 DOI: 10.2147/jir.s499396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/25/2025] [Indexed: 02/11/2025] Open
Abstract
Background Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD), as a common chronic liver condition globally, is experiencing an increasing incidence rate which poses significant health risks. Despite this, the detailed mechanisms underlying the disease's onset and progression remain poorly understood. In this study, we aim to identify effective diagnostic biomarkers for MASLD using microarray data combined with machine learning techniques, which will aid in further understanding the pathogenesis of MASLD. Methods We collected six datasets from the Gene Expression Omnibus (GEO) database, using five of them as training sets and one as a validation set. We employed three machine learning methods-LASSO, SVM, and Random Forest (RF)-to identify hub genes associated with MASLD. These genes were further validated using the external dataset GSE164760. Additionally, functional enrichment analysis, immune infiltration analysis, and immune function analysis were conducted. A TF-miRNA-mRNA network was constructed, and single-cell RNA sequencing was used to determine the distribution of key genes within key cell clusters. Finally, the expression of the key genes was further validated using the palmitic acid-induced AML-12 cell line and the MCD mouse model. Results In this study, through differential gene expression (DEGs) analysis and machine learning techniques, we successfully identified 10 hub genes. Among these, the key gene EGR1 was validated and screened using an external dataset, with an area under the curve (AUC) of 0.882. Enrichment analyses and immune infiltration assessments revealed multiple pathways involving EGR1 in the pathogenesis and progression of MASLD, showing significant correlations with various immune cells. Furthermore, additional cellular experiments and animal model validations confirmed that the expression trends of EGR1 are highly consistent with our analytical findings. Conclusion Our research has confirmed EGR1 as a key gene in MASLD, providing novel insights into the disease's pathogenesis and identifying new therapeutic targets for its treatment.
Collapse
Affiliation(s)
- Xuanlin Wu
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Tao Pan
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Zhihao Fang
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Titi Hui
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Xiaoxiao Yu
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Changxu Liu
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Zihao Guo
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Chang Liu
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| |
Collapse
|
12
|
Zhang J, Hao L, Li S, He Y, Zhang Y, Li N, Hu X. mTOR/HIF-1α pathway-mediated glucose reprogramming and macrophage polarization by Sini decoction plus ginseng soup in ALF. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 137:156374. [PMID: 39798342 DOI: 10.1016/j.phymed.2025.156374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 11/22/2024] [Accepted: 01/03/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND Acute liver failure (ALF) has a high mortality rate, and despite treatment advancements, long-term outcomes remain poor. PURPOSE This study explores the therapeutic targets and pathways of Sini Decoction plus Ginseng Soup (SNRS) in ALF using bioinformatics and network pharmacology, focusing on its impact on macrophage polarization through glucose metabolism reprogramming. The efficacy of SNRS was validated in an LPS/D-GalN-induced ALF model, and its optimal concentration was determined for in vitro macrophage intervention. STUDY DESIGN AND METHODS Differentially expressed genes (DEGs) in HBV-induced and acetaminophen-induced ALF were identified from GEO datasets. The correlation between target gene expression and immune cell infiltration in ALF liver tissue was analyzed. AST, ALT, TNF-α, HMGB1, IL-1β, IL-6, and IL-10 levels were measured, and liver histopathology was assessed. Macrophage polarization was analyzed via immunofluorescence, flow cytometry, and Western blot. Glycolysis-related enzymes and metabolites, including HK2, PFK-1, PKM2, and LDHA, were quantified. Cellular ultrastructure was examined by transmission electron microscopy. RESULTS Five key glycolysis-regulating genes (HK2, CDK1, SOD1, VEGFA, GOT1) were identified, with significant involvement in the HIF-1 signaling pathway. Immune infiltration was markedly higher in ALF liver tissue. SNRS improved survival, reduced ALT/AST levels, alleviated liver injury, and modulated macrophage polarization by decreasing CD86 and increasing CD163 expression. In vitro, SNRS inhibited LPS-induced inflammatory cytokine release, lactate production, p-mTOR/mTOR ratio, and HIF-1α expression. CONCLUSION SNRS modulates macrophage polarization and glucose metabolism reprogramming via the mTOR/HIF-1α pathway, showing promise as a treatment for ALF.
Collapse
Affiliation(s)
- Junli Zhang
- Chengdu University of Traditional Chinese Medicine, No. 37 Shi-er-qiao Road, Chengdu 610075, Sichuan Province, PR China; Department of Infectious Diseases, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Qinghuai District, Nanjing, Jiangsu 210029, PR China
| | - Liyuan Hao
- Chengdu University of Traditional Chinese Medicine, No. 37 Shi-er-qiao Road, Chengdu 610075, Sichuan Province, PR China; Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan Province, PR China
| | - Shenghao Li
- Chengdu University of Traditional Chinese Medicine, No. 37 Shi-er-qiao Road, Chengdu 610075, Sichuan Province, PR China; Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan Province, PR China
| | - Ying He
- Chengdu University of Traditional Chinese Medicine, No. 37 Shi-er-qiao Road, Chengdu 610075, Sichuan Province, PR China; Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan Province, PR China
| | - Yang Zhang
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan Province, PR China
| | - Na Li
- Chengdu University of Traditional Chinese Medicine, No. 37 Shi-er-qiao Road, Chengdu 610075, Sichuan Province, PR China; Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan Province, PR China
| | - Xiaoyu Hu
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan Province, PR China.
| |
Collapse
|
13
|
Feio-Azevedo R, Boesch M, Radenkovic S, van Melkebeke L, Smets L, Wallays M, Boeckx B, Philips G, Prata de Oliveira J, Ghorbani M, Laleman W, Meersseman P, Wilmer A, Cassiman D, van Malenstein H, Triantafyllou E, Sánchez C, Aguilar F, Nevens F, Verbeek J, Moreau R, Arroyo V, Denadai Souza A, Clària J, Lambrechts D, Ghesquière B, Korf H, van der Merwe S. Distinct immunometabolic signatures in circulating immune cells define disease outcome in acute-on-chronic liver failure. Hepatology 2025; 81:509-522. [PMID: 38761406 PMCID: PMC11737128 DOI: 10.1097/hep.0000000000000907] [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: 11/13/2023] [Accepted: 04/07/2024] [Indexed: 05/20/2024]
Abstract
BACKGROUND AND AIMS Acute-on-chronic liver failure (ACLF) is a complication of cirrhosis characterized by multiple organ failure and high short-term mortality. The pathophysiology of ACLF involves elevated systemic inflammation leading to organ failure, along with immune dysfunction that heightens susceptibility to bacterial infections. However, it is unclear how these aspects are associated with recovery and nonrecovery in ACLF. APPROACH AND RESULTS Here, we mapped the single-cell transcriptome of circulating immune cells from patients with ACLF and acute decompensated (AD) cirrhosis and healthy individuals. We further interrogate how these findings, as well as immunometabolic and functional profiles, associate with ACLF-recovery (ACLF-R) or nonrecovery (ACLF-NR). Our analysis unveiled 2 distinct states of classical monocytes (cMons). Hereto, ACLF-R cMons were characterized by transcripts associated with immune and stress tolerance, including anti-inflammatory genes such as RETN and LGALS1 . Additional metabolomic and functional validation experiments implicated an elevated oxidative phosphorylation metabolic program as well as an impaired ACLF-R cMon functionality. Interestingly, we observed a common stress-induced tolerant state, oxidative phosphorylation program, and blunted activation among lymphoid populations in patients with ACLF-R. Conversely, ACLF-NR cMon featured elevated expression of inflammatory and stress response genes such as VIM , LGALS2 , and TREM1 , along with blunted metabolic activity and increased functionality. CONCLUSIONS This study identifies distinct immunometabolic cellular states that contribute to disease outcomes in patients with ACLF. Our findings provide valuable insights into the pathogenesis of ACLF, shedding light on factors driving either recovery or nonrecovery phenotypes, which may be harnessed as potential therapeutic targets in the future.
Collapse
Affiliation(s)
- Rita Feio-Azevedo
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium
| | - Markus Boesch
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium
| | - Silvia Radenkovic
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium
- Metabolomics Expertise Center, Center for Cancer Biology, VIB Center for Cancer Biology, Leuven, Belgium
- Department of Oncology, Metabolomics Expertise Center, KU Leuven, Leuven, Belgium
- Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota, USA
| | - Lukas van Melkebeke
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, UZ Leuven, Leuven, Belgium
| | - Lena Smets
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium
| | - Marie Wallays
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium
| | - Bram Boeckx
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
- VIB Center for Cancer Biology, Leuven, Belgium
| | - Gino Philips
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
- VIB Center for Cancer Biology, Leuven, Belgium
| | - Janaíne Prata de Oliveira
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Mohammad Ghorbani
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium
| | - Wim Laleman
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, UZ Leuven, Leuven, Belgium
| | | | - Alexander Wilmer
- Department of Internal Medicine, UZ Leuven, KU Leuven, Leuven, Belgium
| | - David Cassiman
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium
- Metabolomics Expertise Center, Center for Cancer Biology, VIB Center for Cancer Biology, Leuven, Belgium
- Department of Gastroenterology and Hepatology, UZ Leuven, Leuven, Belgium
| | - Hannah van Malenstein
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, UZ Leuven, Leuven, Belgium
| | - Evangelos Triantafyllou
- Section of Hepatology and Gastroenterology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Cristina Sánchez
- European Foundation for the Study of Chronic Liver Failure, EF-CLIF, EASL-CLIF Consortium and Grifols Chair, Barcelona, Spain
| | - Ferran Aguilar
- European Foundation for the Study of Chronic Liver Failure, EF-CLIF, EASL-CLIF Consortium and Grifols Chair, Barcelona, Spain
| | - Frederik Nevens
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, UZ Leuven, Leuven, Belgium
| | - Jef Verbeek
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, UZ Leuven, Leuven, Belgium
| | - Richard Moreau
- European Foundation for the Study of Chronic Liver Failure, EF-CLIF, EASL-CLIF Consortium and Grifols Chair, Barcelona, Spain
- Centre de Recherche sur l’Inflammation (CRI) UMRS1149, Université de Paris Cité, Service d’Hépatologie, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris, Clichy, France
| | - Vicente Arroyo
- European Foundation for the Study of Chronic Liver Failure, EF-CLIF, EASL-CLIF Consortium and Grifols Chair, Barcelona, Spain
| | | | - Joan Clària
- European Foundation for the Study of Chronic Liver Failure, EF-CLIF, EASL-CLIF Consortium and Grifols Chair, Barcelona, Spain
- Hospital Clínic-IDIBAPS, CIBERehd, Universitat de Barcelona, Barcelona, Spain
| | - Diether Lambrechts
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
- VIB Center for Cancer Biology, Leuven, Belgium
| | - Bart Ghesquière
- Metabolomics Expertise Center, Center for Cancer Biology, VIB Center for Cancer Biology, Leuven, Belgium
- Department of Oncology, Metabolomics Expertise Center, KU Leuven, Leuven, Belgium
| | - Hannelie Korf
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium
| | - Schalk van der Merwe
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, UZ Leuven, Leuven, Belgium
| |
Collapse
|
14
|
Triantafyllou E, Gudd CLC, Possamai LA. Immune-mediated liver injury from checkpoint inhibitors: mechanisms, clinical characteristics and management. Nat Rev Gastroenterol Hepatol 2025; 22:112-126. [PMID: 39663461 DOI: 10.1038/s41575-024-01019-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/05/2024] [Indexed: 12/13/2024]
Abstract
Immunotherapy has changed the treatment landscape for patients with cancer in the past decade. Immune checkpoint inhibitor (ICI)-based therapies have proven effective in a range of malignancies, including liver and gastrointestinal cancers, but they can cause diverse off-target organ toxicities. With the increasingly wider application of these drugs, immune-mediated liver injury from ICIs has become a commonly encountered challenge in clinical hepatology and gastroenterology. In this Review, we discuss the evidence from human and animal studies on the immunological mechanisms of immune-mediated liver injury from ICIs and summarize its clinical features and practical considerations for its management.
Collapse
Affiliation(s)
- Evangelos Triantafyllou
- Section of Hepatology and Gastroenterology, Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom.
| | - Cathrin L C Gudd
- Section of Hepatology and Gastroenterology, Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Lucia A Possamai
- Section of Hepatology and Gastroenterology, Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom.
- Liver and Antiviral Unit, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom.
| |
Collapse
|
15
|
Yu S, Pei S, Zhang M, Gao S, Chen J, Duan L, Hu E, Wang Y, Huang Y. PKM2-mediated STAT3 phosphorylation promotes acute liver failure via regulating NLRP3-dependent pyroptosis. Commun Biol 2024; 7:1694. [PMID: 39722076 DOI: 10.1038/s42003-024-07227-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 11/07/2024] [Indexed: 12/28/2024] Open
Abstract
Acute liver failure (ALF) is a life-threatening clinical syndrome characterized by high-grade inflammation and multi-organ failure. Our previous study shows that targeting the M2 isoform of pyruvate kinase (PKM2) to inhibit macrophage inflammation may be a promising strategy for ALF treatment. however, the mechanism by which PKM2 regulates the inflammatory response is unclear. Here we demonstrate that PKM2 contributes to ALF by modulating NLRP3-mediated pyroptosis activation in liver macrophages. The specific knockout of PKM2 in myeloid cells reduces mortality and alleviates hepatic injury in D-galactosamine/LPS-induced ALF mice. Single-cell transcriptome analysis suggests that NLRP3 inflammasome activation of macrophages involves in ALF, knockout of PKM2 in macrophages reduces the expression of NLRP3, and activation of pyroptosis. Pharmacological inhibition of the PKM2 nuclear translocation, but not glycolytic activity, protects mice from ALF. Pharmacological and genetic inhibition of PKM2 attenuates NLRP3-mediated pyroptosis activation and consequently reduces the release of IL-1β and IL-18 by macrophages. Mechanistically, PKM2 translocates into the nucleus and combines with STAT3, enhancing its phosphorylation and recruitment to the NLRP3 promoter region, thereby increasing NLRP3 expression. This work defines PKM2 acts as an important nonmetabolic regulator of NLRP3 that modulates pyroptosis activation in macrophages and guides future therapeutic strategies development for ALF.
Collapse
Affiliation(s)
- Songman Yu
- Department of Infectious Diseases, Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| | - Siya Pei
- Department of Blood Transfusion, Xiangya Hospital, Central South University, Changsha, China
| | - Min Zhang
- Department of Infectious Diseases, Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| | - Shang Gao
- Department of Infectious Diseases, Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| | - Jun Chen
- Department of Infectious Diseases, Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| | - Lihua Duan
- Department of Infectious Diseases, Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| | - En Hu
- Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Yang Wang
- Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, China.
| | - Yan Huang
- Department of Infectious Diseases, Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
16
|
Zhang J, Li N, Hu X. Metabolic Reprograming of Macrophages: A New Direction in Traditional Chinese Medicine for Treating Liver Failure. J Immunol Res 2024; 2024:5891381. [PMID: 39741958 PMCID: PMC11688140 DOI: 10.1155/jimr/5891381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 09/03/2024] [Accepted: 11/28/2024] [Indexed: 01/03/2025] Open
Abstract
Acute liver failure (ALF) is a fulminant clinical syndrome that usually leads to multiple organ failure and high mortality. Macrophages play a crucial role in the initiation, development, and recovery of ALF. Targeting macrophages through immunotherapy holds significant promise as a therapeutic strategy. These cells exhibit remarkable plasticity, enabling them to differentiate into various subtypes based on changes in their surrounding microenvironment. M1-type macrophages are associated with a pro-inflammatory phenotype and primarily rely predominantly on glycolysis. In contrast, M2-type macrophages, which are characterized by anti-inflammatory phenotype, predominantly obtain their energy from oxidative phosphorylation (OXPHOS) and fatty acid oxidation (FAO). Shifting macrophage metabolism from glycolysis to OXPHOS inhibits M1 macrophage activation and promotes M2 macrophage activation, thereby exerting anti-inflammatory and reparative effects. This study elucidates the relationship between macrophage activation and glucose metabolism reprograming from an immunometabolism perspective. A comprehensive literature review revealed that several signaling pathways may regulate macrophage polarization through energy metabolism, including phosphatidyl-inositol 3-kinase/protein kinase B (PI3K/AKT), mammalian target of rapamycin (mTOR)/hypoxia-inducible factor 1α (HIF-1α), nuclear factor-κB (NF-κB), and AMP-activated protein kinase (AMPK), which exhibit crosstalk with one another. Additionally, we systematically reviewed several traditional Chinese medicine (TCM) monomers that can modulate glucose metabolism reprograming and influence the polarization states of M1 and M2 macrophages. This review aimed to provide valuable insights that could contribute to the development of new therapies or drugs for ALF.
Collapse
Affiliation(s)
- Junli Zhang
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Na Li
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaoyu Hu
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
17
|
Cheng K, Liu K, Liu S, Zhao Y, Wang Q. IGF2BP3 regulates macrophage-induced inflammation and liver damage in acute-on-chronic liver failure via the RORα-NF-κB signaling axis. Int Immunopharmacol 2024; 142:113030. [PMID: 39232362 DOI: 10.1016/j.intimp.2024.113030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/23/2024] [Accepted: 08/25/2024] [Indexed: 09/06/2024]
Abstract
Acute-on-chronic liver failure (ACLF) is a severe condition characterized by high mortality rates, and macrophage-mediated inflammation plays a critical role in its progression. Our previous research has indicated the involvement of the RNA-binding protein IGF2BP3 in the pathogenesis of ACLF. However, the underlying molecular mechanisms contributing to this damage require further elucidation. Initially, we observed heightened expression of pro-inflammatory cytokines and macrophage activation in both ACLF patients and a mouse model induced by D-GalN/LPS. Subsequent loss-of-function experiments targeting IGF2BP3 revealed that the knockdown of IGF2BP3 potentially confers hepatoprotection by mitigating macrophage-induced inflammation. Further investigation using RNA Immunoprecipitation (RIP) assays and dual luciferase reporter assays confirmed that RORα is a target protein of the RNA-binding protein IGF2BP3. Importantly, depletion of RORα was found to significantly increase liver damage and inflammation by modulating the NF-κB signaling pathway. In conclusion, our findings underscore the crucial role of IGF2BP3 in mediating liver damage induced by activated macrophages in ACLF, which is regulated by the RORα-NF-κB signaling pathway. These discoveries offer novel insights into the pathogenesis and potential therapeutic targets for ACLF.
Collapse
Affiliation(s)
- Ke Cheng
- Department of Transplantation, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China; Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, Hunan 410013, China
| | - Kai Liu
- Department of Transplantation, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China; Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, Hunan 410013, China
| | - Shu Liu
- Department of Transplantation, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China; Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, Hunan 410013, China
| | - Yujun Zhao
- Department of Transplantation, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China; Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, Hunan 410013, China
| | - Qiang Wang
- Department of Transplantation, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China; Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, Hunan 410013, China.
| |
Collapse
|
18
|
Zhou JY, Lu YN, Shen XY, Quan YZ, Lu JM, Jin GN, Liu YM, Zhang SH, Xu GH, Xu X, Piao LX. Coixol mitigates Toxoplasma gondii infection-induced liver injury by inhibiting the Toxoplasma gondii HSP70/TLR4/NF-κB signaling pathway in hepatic macrophages. JOURNAL OF ETHNOPHARMACOLOGY 2024; 335:118694. [PMID: 39147001 DOI: 10.1016/j.jep.2024.118694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 07/12/2024] [Accepted: 08/10/2024] [Indexed: 08/17/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Coix seed, the dry mature seed kernel of the gramineous plant coix (Coix lacryma-jobi L. var. ma-yuen Stapf), is widely consumed as a traditional Chinese medicine and functional food in China and South Korea. We have previously demonstrated the protective effect of coixol, a polyphenolic compound extracted from coix, against Toxoplasma gondii (T. gondii) infection-induced lung injury. However, the protective effect of coixol on hepatic injury induced by T. gondii infection have not yet been elucidated. AIM OF THE STUDY This study explores the impact of coixol on T. gondii infection-induced liver injury and elucidates the underlying molecular mechanisms. MATERIALS AND METHODS Female BALB/c mice and Kupffer cells (KCs) were employed to establish an acute T. gondii infection model in vivo and an inflammation model in vitro. The study examined coixol's influence on the T. gondii-derived heat shock protein 70 (T.g.HSP70)/toll-like receptor 4 (TLR4)/nuclear factor (NF)-κB signaling pathway in T. gondii-infected liver macrophages. Furthermore, a co-culture system of KCs and NCTC-1469 hepatocytes was developed to observe the impact of liver macrophages infected with T. gondii on hepatocyte injury. RESULTS Coixol notably inhibited the proliferation of tachyzoites and the expression of T.g.HSP70 in mouse liver and KCs, and attenuated pathological liver injury. Moreover, coixol decreased the production of high mobility group box 1, tumor necrosis factor-α, and inducible nitric oxide synthase by suppressing the TLR4/NF-κB signaling pathway in vitro and in vivo. Coixol also mitigated KCs-mediated hepatocyte injury. CONCLUSIONS Coixol protects against liver injury caused by T. gondii infection, potentially by diminishing hepatocyte injury through the suppression of the inflammatory cascade mediated by the T.g.HSP70/TLR4/NF-κB signaling pathway in KCs. These findings offer new perspectives for developing coixol as a lead compound for anti-T. gondii drugs.
Collapse
Affiliation(s)
- Jin-Yi Zhou
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Yu-Nan Lu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Xin-Yu Shen
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Yan-Zhu Quan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Jing-Mei Lu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Guang-Nan Jin
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Yi-Ming Liu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Si-Hui Zhang
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Guang-Hua Xu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China.
| | - Xiang Xu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China.
| | - Lian-Xun Piao
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China.
| |
Collapse
|
19
|
Sun F, Wang J, Ji X, Wang Z, Gao S, Wang K. CCL25 contributes to the pathogenesis of D-Gal/LPS-induced acute liver failure. J Gastroenterol Hepatol 2024; 39:2880-2891. [PMID: 39233339 DOI: 10.1111/jgh.16732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/14/2024] [Accepted: 08/21/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND AND AIM Acute liver failure (ALF) is a fatal clinical syndrome of severe hepatic dysfunction. Chemokines promote liver diseases by recruiting and activating immune cells. We aimed to investigate the role of C-C chemokine ligand 25 (CCL25) in ALF. METHODS An ALF mouse model induced by D-galactosamine/lipopolysaccharide was evaluated through liver hematoxylin and eosin staining and serum transaminase and cytokine measurement. CCL25 expression in serum was analyzed by ELISA and in liver by immunohistochemical staining and western blot. C-C chemokine receptor 9 (CCR9)-expressing cells in the liver were identified by immunofluorescence staining. The effects of anti-CCL25 on ALF were evaluated in vivo. Cytokine expression and migration of CCL25-stimulated RAW264.7 macrophages were studied. We also investigated the role of anti-CCL25 and BMS-345541, an NF-κB signaling inhibitor, in vitro. NF-κB activation was assessed via western blot, and p65 nuclear translocation was detected using cellular immunofluorescence. RESULTS ALF mice showed severe histological damage and high serum levels of aminotransferase and inflammatory cytokines. Elevated CCL25 and NF-κB activation was observed in vivo. CCR9 was expressed on macrophages in ALF mouse liver. ALF was suppressed after anti-CCL25 treatment, with significant NF-κB inhibition. In vitro, CCL25 induced strong migration and cytokine release in RAW264.7 macrophages, which were eliminated by anti-CCL25 and BMS-345541. Furthermore, the NF-κB activation and p65 nuclear translocation induced by CCL25 were also inhibited by anti-CCL25 and BMS-345541. CONCLUSION CCL25 contributes to ALF development by inducing macrophage-mediated inflammation via activation of the NF-κB signaling.
Collapse
Affiliation(s)
- Fei Sun
- Department of Hepatology, Qilu Hospital (Qingdao) of Shandong University, Qingdao, China
| | - Jingwei Wang
- Department of Hepatology, Qilu Hospital (Qingdao) of Shandong University, Qingdao, China
| | - Xiangfen Ji
- Department of Hepatology, Qilu Hospital (Qingdao) of Shandong University, Qingdao, China
| | - Zhenli Wang
- Department of Hepatology, Qilu Hospital (Qingdao) of Shandong University, Qingdao, China
| | - Shuai Gao
- Department of Hepatology, 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
| |
Collapse
|
20
|
Auzinger G. Basic concepts in the management of Acute Liver Failure. Best Pract Res Clin Gastroenterol 2024; 73:101960. [PMID: 39709220 DOI: 10.1016/j.bpg.2024.101960] [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: 09/02/2024] [Accepted: 10/29/2024] [Indexed: 12/23/2024]
Affiliation(s)
- Georg Auzinger
- King's College Hospital, Liver Intensive Care Unit, London, UK; Cleveland Clinic London, Department of Critical Care, London, UK.
| |
Collapse
|
21
|
Xu FF, Li ZC, Zhang WJ, Li Q, Li DJ, Meng HB, Shen FM, Fu H. Activation of α7 Nicotinic Acetylcholine Receptors Inhibits Hepatic Necroptosis and Ameliorates Acute Liver Injury in Mice. Anesthesiology 2024; 141:1119-1138. [PMID: 39186677 DOI: 10.1097/aln.0000000000005206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
BACKGROUND Acute liver injury is a disease characterized by severe liver dysfunction, caused by significant infiltration of immune cells and extensive cell death with a high mortality. Previous studies demonstrated that the α7 nicotinic acetylcholine receptor (α7nAChR) played a crucial role in various liver diseases. The hypothesis of this study was that activating α7nAChR could alleviate acute liver injury and investigate its possible mechanisms. METHODS Acute liver injury was induced by intraperitoneal injection of lipopolysaccharide (LPS)/D-galactosamine (D-Gal) in wild type, α7nAChR knockout (α7nAChR-/-) and stimulator of interferon gene (STING) mutation (Stinggt/gt) mice in the presence or absence of a pharmacologic selective α7nAChR agonist (PNU-282987). The effects of α7nAChR on hepatic injury, inflammatory response, mitochondrial damage, necroptosis, and infiltration of immune cells during acute liver injury were assessed. RESULTS The expression of α7nAChR in liver tissue was increased in LPS/D-Gal-induced acute liver injury mice. Compared to the age-matched wild-type mice, α7nAChR deficiency decreased the survival rate, exacerbated the hepatic injury accompanied with enhanced inflammatory response and oxidative stress, and aggravated hepatic mitochondrial damage and necroptosis. Conversely, pharmacologic activation of α7nAChR by PNU-282987 displayed the opposite trends. Furthermore, PNU-282987 significantly reduced the proportion of infiltrating monocyte-derived macrophages (CD45+CD11bhiF4/80int), M1 macrophages (CD45+CD11b+F4/80+CD86hiCD163low), and Ly6Chi monocytes (CD45+CD11b+MHC [major histocompatibility complex] ⅡlowLy6Chi), but increased the resident Kupffer cells (CD45+CD11bintF4/80hiTIM4hi) in the damaged hepatic tissues caused by LPS/D-Gal. Interestingly, α7nAChR deficiency promoted the STING signaling pathway under LPS/D-Gal stimulation, while PNU-282987 treatment significantly prevented its activation. Finally, it was found that Sting mutation abolished the protective effects against hepatic injury by activating α7nAChR. CONCLUSIONS The authors' study revealed that activating α7nAChR could protect against LPS/D-Gal-induced acute liver injury by inhibiting hepatic inflammation and necroptosis possibly via regulating immune cells infiltration and inhibiting STING signaling pathway. EDITOR’S PERSPECTIVE
Collapse
Affiliation(s)
- Fang-Fang Xu
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zi-Chen Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wen-Jing Zhang
- Department of Pharmacy, Inner Mongolia Autonomous Region People's Hospital, Hohhot, China
| | - Qiao Li
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Dong-Jie Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hong-Bo Meng
- Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Fu-Ming Shen
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hui Fu
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
22
|
Niemietz P, Peiseler M, Kohlhepp M, Horn P, Matchett K, Wang Y, Haas L, Zhang T, Bruneau A, Guillot A, Berger H, Liepelt A, Warzecha K, Demske C, Möckel D, Lammers T, Henderson N, Heymann F, Tacke F. C-C chemokine receptor type 7 (CCR7) regulates hepatic CD8 + T cell homeostasis and response to acute liver injury. Hepatology 2024; 80:1104-1119. [PMID: 38231043 DOI: 10.1097/hep.0000000000000757] [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: 08/21/2023] [Accepted: 12/20/2023] [Indexed: 01/18/2024]
Abstract
BACKGROUND AND AIMS Acute liver failure (ALF) is a rare but life-threatening condition, and DILI, particularly acetaminophen toxicity, is the leading cause of ALF. Innate immune mechanisms further perpetuate liver injury, while the role of the adaptive immune system in DILI-related ALF is unclear. APPROACH AND RESULTS We analyzed liver tissue from 2 independent patient cohorts with ALF and identified hepatic T cell infiltration as a prominent feature in human ALF. CD8 + T cells were characterized by zonation toward necrotic regions and an activated gene expression signature. In murine acetaminophen-induced liver injury, intravital microscopy revealed zonation of CD8 + but not CD4 + T cells at necrotic areas. Gene expression analysis exposed upregulated C-C chemokine receptor 7 (CCR7) and its ligand CCL21 in the liver as well as a broadly activated phenotype of hepatic CD8 + T cells. In 2 mouse models of ALF, Ccr7-/- mice had significantly aggravated early-phase liver damage. Functionally, CCR7 was not involved in the recruitment of CD8 + T cells, but regulated their activation profile potentially through egress to lymphatics. Ccr7-/- CD8 + T cells were characterized by elevated expression of activation, effector, and exhaustion profiles. Adoptive transfer revealed preferential homing of CCR7-deficient CD8 + T cells to the liver, and depletion of CD8 + T cells attenuated liver damage in mice. CONCLUSIONS Our study demonstrates the involvement of the adaptive immune system in ALF in humans and mice. We identify the CCR7-CCL21 axis as an important regulatory pathway, providing downstream protection against T cell-mediated liver injury.
Collapse
Affiliation(s)
- Patricia Niemietz
- Department of Medicine III, RWTH-University Hospital Aachen, Aachen, Germany
| | - Moritz Peiseler
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Marlene Kohlhepp
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Paul Horn
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Kylie Matchett
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Yuting Wang
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Leon Haas
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Tianjiao Zhang
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Alix Bruneau
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Adrien Guillot
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Hilmar Berger
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Anke Liepelt
- Department of Medicine III, RWTH-University Hospital Aachen, Aachen, Germany
| | - Klaudia Warzecha
- Department of Medicine III, RWTH-University Hospital Aachen, Aachen, Germany
| | - Catharina Demske
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Diana Möckel
- Department of Nanomedicine and Theranostics, Institute for Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Neil Henderson
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Felix Heymann
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Frank Tacke
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| |
Collapse
|
23
|
Zhang D, Shi C, Wang Y, Guo J, Gong Z. Metabolic Dysregulation and Metabolite Imbalances in Acute-on-chronic Liver Failure: Impact on Immune Status. J Clin Transl Hepatol 2024; 12:865-877. [PMID: 39440217 PMCID: PMC11491507 DOI: 10.14218/jcth.2024.00203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/26/2024] [Accepted: 09/04/2024] [Indexed: 10/25/2024] Open
Abstract
Liver failure encompasses a range of severe clinical syndromes resulting from the deterioration of liver function, triggered by factors both within and outside the liver. While the definition of acute-on-chronic liver failure (ACLF) may vary by region, it is universally recognized for its association with multiorgan failure, a robust inflammatory response, and high short-term mortality rates. Recent advances in metabolomics have provided insights into energy metabolism and metabolite alterations specific to ACLF. Additionally, immunometabolism is increasingly acknowledged as a pivotal mechanism in regulating immune cell functions. Therefore, understanding the energy metabolism pathways involved in ACLF and investigating how metabolite imbalances affect immune cell functionality are crucial for developing effective treatment strategies for ACLF. This review methodically examined the immune and metabolic states of ACLF patients and elucidated how alterations in metabolites impact immune functions, offering novel perspectives for immune regulation and therapeutic management of liver failure.
Collapse
Affiliation(s)
- Danmei Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Chunxia Shi
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yukun Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jin Guo
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zuojiong Gong
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
24
|
Lu J, Tu Z, Zhang Z, Wang S, Liu Z, Lu X, Zhang J, Luo D. The rate of change in clinical indicators can predict the progression of hepatitis B virus-related acute-on-chronic preliver failure. Medicine (Baltimore) 2024; 103:e40246. [PMID: 39470556 PMCID: PMC11521059 DOI: 10.1097/md.0000000000040246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 06/10/2024] [Accepted: 10/02/2024] [Indexed: 10/30/2024] Open
Abstract
The objective of this study was to investigate the predictors and predictive model construction of the progression of HBV-Pre.Acute-on-chronic liver failure (ACLF), a total of 133 patients with HBV-Pre.ACLF was divided into the progressive group (52 patients) and the recovery group (81 patients) according to whether they progressed to ACLF or not. The clinical parameters N%, L%, PLT, ALT, TBiL, ALB, Cre, Na, NH3, CRP, AFP, prothrombin time (PT), international normalized ratio (INR), FIB, and their rate of change at baseline were analyzed in the 2 groups. The independent risk factors for HBV-Pre.ACLF progression was found by univariate and multivariate analyses, and a predictive model was constructed. The clinical parameters ALB, FIB, Na, combined alprostadil treatment and MELD, and MELD-Na scores at baseline were significantly different between the 2 groups (P <.05), while ALT, TBiL, Cre, CHE, NH3, N%, L%, PLT, INR, and PT were not significantly different (P >.05). The change rates of Na, CHE, PT, FIB, CRP, Cre, PLT, and the ratio after to before of N% were significantly different between the 2 groups (P <.05), while the change rates of ALT, TBIL, NH3, AFP, L%, and the ratio after to before of INR were not significantly different between the 2 groups (P >.05). Univariate and multivariate analyses showed that baseline ALB, Na, FIB, combined alprostadil therapy and the rate of change of Na and PLT were protective factors for disease progression, and the rate of change of PT, CRP, and the ratio after to before of N% were independent risk factors for disease progression. The novel model was LogitP = -6.051 + 4.049×ΔPT + 0.626×ΔCRP + 4.527×the ratio after to before N% and its area under the curve was 0.944 (95% confidence interval: 0.900-0.988) predicting progression of HBV-Pre.ACLF, and the best cutoff value was -0.22. The patients with a higher logitP score (> -0.22) had an increased risk for progression to ACLF (P <.05). The novel model logitP shows good predictive value for the disease progression of HBV-Pre.ACLF.
Collapse
Affiliation(s)
- Jun Lu
- Department of Intensive Care Unit, Infectious Disease Hospital Affiliated to Nanchang University, Nanchang, Jiangxi, China
| | - Zhihui Tu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zhen Zhang
- Department of Intensive Care Unit, Infectious Disease Hospital Affiliated to Nanchang University, Nanchang, Jiangxi, China
| | - Shumei Wang
- Department of Intensive Care Unit, Infectious Disease Hospital Affiliated to Nanchang University, Nanchang, Jiangxi, China
| | - Zhuoqi Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xiaohui Lu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jun Zhang
- Department of Intensive Care Unit, Infectious Disease Hospital Affiliated to Nanchang University, Nanchang, Jiangxi, China
| | - Daya Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
25
|
Cavazza A, Triantafyllou E, Savoldelli R, Mujib S, Jerome E, Trovato FM, Artru F, Sheth R, Huang XH, Ma Y, Dazzi F, Pirani T, Antoniades CG, Lee WM, McPhail MJ, Karvellas CJ. Macrophage activation markers are associated with infection and mortality in patients with acute liver failure. Liver Int 2024; 44:1900-1911. [PMID: 38588014 PMCID: PMC11466005 DOI: 10.1111/liv.15928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 03/18/2024] [Accepted: 03/21/2024] [Indexed: 04/10/2024]
Abstract
BACKGROUND AND AIMS Acute liver failure is a multisystem disorder with a high mortality and frequent need for emergency liver transplantation. Following massive innate immune system activation, soluble markers of macrophage activation are released during liver damage and their association with disease severity and prognosis requires exploration. METHODS Patients ALF from the United States Acute Liver Failure Study Group (USALFSG, n = 224) and King's College Hospital (n = 40) together with healthy controls (HC, n = 50) were recruited. Serum from early (Days 1-3) and late (>Day 3) time points were analysed for MAMs by enzyme-linked immunosorbent assay correlated to markers of illness severity and 21-day spontaneous survival. Surface expression phenotyping was performed via Flow Cytometry on CD14+ monocytes. RESULTS All MAMs serum concentrations were significantly higher in ALF compared to controls (p < .0001). sCD206 concentration was higher in early and late stages of the disease in patients with bacteraemia (p = .002) and infection in general (p = .006). In MELD-adjusted multivariate modelling, sCD206 and sCD163 were independently associated with mortality. CD14+ monocyte expression of CD206 (p < .001) was higher in patients with ALF compared with controls and correlated with SOFA score (p = .018). sCD206 was independently validated as a predictor of infection in an external cohort. CONCLUSIONS sCD206 is increased in serum of ALF patients with infections and poor outcome and is upregulated on CD14+ monocytes. Later measurements of sCD163 and sCD206 during the evolution of ALF have potential as mechanistic predictors of mortality. sCD206 should be explored as a biomarker of sepsis and mortality in ALF.
Collapse
Affiliation(s)
- Anna Cavazza
- Department of Inflammation Biology, School of Inflammation and Microbial Science, Institute of Liver StudiesKing's College LondonLondonUK
- Liver Intensive Therapy UnitInstitute of Liver Studies, King's College HospitalLondonUK
| | - Evangelos Triantafyllou
- Section of Hepatology and Gastroenterology, Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
| | - Roberto Savoldelli
- School of Cardiovascular and Metabolic Medicine and ScienceKing's College LondonLondonUK
| | - Salma Mujib
- Department of Inflammation Biology, School of Inflammation and Microbial Science, Institute of Liver StudiesKing's College LondonLondonUK
- Liver Intensive Therapy UnitInstitute of Liver Studies, King's College HospitalLondonUK
| | - Ellen Jerome
- Department of Inflammation Biology, School of Inflammation and Microbial Science, Institute of Liver StudiesKing's College LondonLondonUK
- Liver Intensive Therapy UnitInstitute of Liver Studies, King's College HospitalLondonUK
| | - Francesca M. Trovato
- Department of Inflammation Biology, School of Inflammation and Microbial Science, Institute of Liver StudiesKing's College LondonLondonUK
- Liver Intensive Therapy UnitInstitute of Liver Studies, King's College HospitalLondonUK
| | - Florent Artru
- Department of Inflammation Biology, School of Inflammation and Microbial Science, Institute of Liver StudiesKing's College LondonLondonUK
- Liver Intensive Therapy UnitInstitute of Liver Studies, King's College HospitalLondonUK
| | - Roosey Sheth
- Department of Inflammation Biology, School of Inflammation and Microbial Science, Institute of Liver StudiesKing's College LondonLondonUK
- Liver Intensive Therapy UnitInstitute of Liver Studies, King's College HospitalLondonUK
| | - Xiao Hong Huang
- Department of Inflammation Biology, School of Inflammation and Microbial Science, Institute of Liver StudiesKing's College LondonLondonUK
| | - Yun Ma
- Department of Inflammation Biology, School of Inflammation and Microbial Science, Institute of Liver StudiesKing's College LondonLondonUK
| | - Francesco Dazzi
- School of Cardiovascular and Metabolic Medicine and ScienceKing's College LondonLondonUK
| | - Tasneem Pirani
- Department of Inflammation Biology, School of Inflammation and Microbial Science, Institute of Liver StudiesKing's College LondonLondonUK
- Liver Intensive Therapy UnitInstitute of Liver Studies, King's College HospitalLondonUK
| | - Charalambos G. Antoniades
- Department of Inflammation Biology, School of Inflammation and Microbial Science, Institute of Liver StudiesKing's College LondonLondonUK
- Liver Intensive Therapy UnitInstitute of Liver Studies, King's College HospitalLondonUK
| | - William M. Lee
- Division of Digestive and Liver DiseasesUT Southwestern Medical CenterDallasTexasUSA
| | - Mark J. McPhail
- Department of Inflammation Biology, School of Inflammation and Microbial Science, Institute of Liver StudiesKing's College LondonLondonUK
- Liver Intensive Therapy UnitInstitute of Liver Studies, King's College HospitalLondonUK
| | - Constantine J. Karvellas
- Division of Gastroenterology (Liver Unit), Department of Critical Care MedicineUniversity of AlbertaEdmontonCanada
| | | |
Collapse
|
26
|
Chen J, Zhang Q, Xu W, Li Z, Chen X, Luo Q, Wang D, Peng L. Baicalein upregulates macrophage TREM2 expression via TrKB-CREB1 pathway to attenuate acute inflammatory injury in acute-on-chronic liver failure. Int Immunopharmacol 2024; 139:112685. [PMID: 39047449 DOI: 10.1016/j.intimp.2024.112685] [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: 02/07/2024] [Revised: 06/06/2024] [Accepted: 07/11/2024] [Indexed: 07/27/2024]
Abstract
OBJECTIVE Acute-on-chronic liver failure (ACLF) is a syndrome characterized by a high short-term mortality rate, and effective interventions are still lacking. This study aims to investigate whether the small molecule baicalein can mitigate ACLF and elucidate the molecular mechanisms. METHODS The ACLF mouse model was induced through chronic liver injury using carbon tetrachloride, followed by acute inflammation induction with lipopolysaccharide (LPS). Baicalein was administered through intraperitoneal injection to explore its therapeutic effects. In vitro experiments utilized the iBMDM macrophage cell line to investigate the underlying mechanisms. Peripheral blood was collected from clinical ACLF patients for validation. RESULTS In the LPS-induced ACLF mouse model, baicalein demonstrated a significant reduction in acute inflammation and liver damage, as evidenced by histopathological evaluation, liver function analysis, and inflammatory marker measurements. Transcriptomic analysis, coupled with molecular biology experiments, uncovered that baicalein exerts its effects in ACLF by activating the TrKB-CREB1 signaling axis to upregulate the surface expression of the TREM2 receptor on macrophages. This promotes M2 macrophage polarization and activates efferocytosis, thereby inhibiting inflammation and alleviating liver damage. Furthermore, we observed a substantial negative correlation between postoperative peripheral blood plasma soluble TREM2 (sTREM2) levels and inflammation, as well as adverse outcomes in clinical ACLF patients. CONCLUSION Baicalein plays a protective role in ACLF by enhancing the surface expression of the TREM2 receptor on macrophages, leading to the suppression of inflammation, mitigation of liver damage, and a reduction in mortality. Additionally, plasma sTREM2 emerges as a critical indicator for predicting adverse outcomes in ACLF patients.
Collapse
Affiliation(s)
- Jia Chen
- Department of Infectious Diseases and Guangdong Key Laboratory of Liver Disease Research, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qiongchi Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China; Department of Orthopedics, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Wenxiong Xu
- Department of Infectious Diseases and Guangdong Key Laboratory of Liver Disease Research, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhipeng Li
- Department of Infectious Diseases and Guangdong Key Laboratory of Liver Disease Research, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiyao Chen
- Department of Infectious Diseases and Guangdong Key Laboratory of Liver Disease Research, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qiumin Luo
- Department of Infectious Diseases and Guangdong Key Laboratory of Liver Disease Research, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Dong Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China; Department of Orthopedics, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.
| | - Liang Peng
- Department of Infectious Diseases and Guangdong Key Laboratory of Liver Disease Research, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
27
|
Wei J, Sun Y, Wang H, Zhu T, Li L, Zhou Y, Liu Q, Dai Z, Li W, Yang T, Wang B, Zhu C, Shen X, Yao Q, Song G, Zhao Y, Pei H. Designer cellular spheroids with DNA origami for drug screening. SCIENCE ADVANCES 2024; 10:eado9880. [PMID: 39028810 PMCID: PMC11259176 DOI: 10.1126/sciadv.ado9880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 06/14/2024] [Indexed: 07/21/2024]
Abstract
Current in vitro models struggle to balance the complexity of human diseases with suitability for large-scale drug tests. While 3D cultures simulate human tissues, they lack cellular intricacy, and integrating these models with high-throughput drug screening remains a challenge. Here, we introduce a method that uses self-assembling nucleic acid nanostructures decorated living cells, termed NACs, to create spheroids with a customizable 3D layout. To demonstrate its uniqueness, our method effectively creates designer 3D spheroids by combining parenchymal cells, stromal cells, and immune cells, leading to heightened physiological relevance and detailed modeling of complex chronic diseases and immune-stromal interactions. Our approach achieves a high level of biological fidelity while being standardized and straightforward to construct with the potential for large-scale drug discovery applications. By merging the precision of DNA nanotechnology with advanced cell culture techniques, we are streamlining human-centric models, striking a balance between complexity and standardization, to boost drug screening efficiency.
Collapse
Affiliation(s)
- Jiayi Wei
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Institute of Liver Diseases, Shanghai 200032, China
| | - Yueyang Sun
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, Shanghai Center of Brain-inspired Intelligent Materials and Devices, East China Normal University, Shanghai 200241, China
| | - Heming Wang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Institute of Liver Diseases, Shanghai 200032, China
| | - Tong Zhu
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, Shanghai Center of Brain-inspired Intelligent Materials and Devices, East China Normal University, Shanghai 200241, China
| | - Li Li
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, Shanghai Center of Brain-inspired Intelligent Materials and Devices, East China Normal University, Shanghai 200241, China
| | - Ying Zhou
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, Shanghai Center of Brain-inspired Intelligent Materials and Devices, East China Normal University, Shanghai 200241, China
| | - Quan Liu
- Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun 130117, China
| | - Zhen Dai
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Wenjuan Li
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Taihua Yang
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200001, China
| | - Bingmei Wang
- Changchun University of Chinese Medicine, Changchun 130117, China
| | - Changfeng Zhu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Institute of Liver Diseases, Shanghai 200032, China
| | - Xizhong Shen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Institute of Liver Diseases, Shanghai 200032, China
| | - Qunyan Yao
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Institute of Liver Diseases, Shanghai 200032, China
- Shanghai Geriatric Medical Center, Shanghai 201104, China
- Department of Gastroenterology and Hepatology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen 361015, China
| | - Guangqi Song
- Joint Laboratory of Biomaterials and Translational Medicine, Puheng Technology, Suzhou 215000, China
| | - Yicheng Zhao
- Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun 130117, China
- China-Japan Union Hospital of Jilin University, 130012 Changchun, Jilin, China
| | - Hao Pei
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, Shanghai Center of Brain-inspired Intelligent Materials and Devices, East China Normal University, Shanghai 200241, China
| |
Collapse
|
28
|
Wang K, Zhu Y, Li M, Yang Y, Zuo D, Sheng J, Zhang X, Wang W, Zhou P, Feng M. Genetically Modified Hepatocytes Targeting Bilirubin and Ammonia Metabolism for the Construction of Bioartificial Liver System. Biomater Res 2024; 28:0043. [PMID: 39011520 PMCID: PMC11246981 DOI: 10.34133/bmr.0043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 05/08/2024] [Indexed: 07/17/2024] Open
Abstract
Acute liver failure (ALF) is a complex syndrome that impairs the liver's function to detoxify bilirubin, ammonia, and other toxic metabolites. Bioartificial liver (BAL) aims to help ALF patients to pass through the urgent period by temporarily undertaking the liver's detoxification functions and promoting the recovery of the injured liver. We genetically modified the hepatocellular cell line HepG2 by stably overexpressing genes encoding UGT1A1, OATP1B1, OTC, ARG1, and CPS1. The resulting SynHeps-II cell line, encapsulated by Cytopore microcarriers, dramatically reduced the serum levels of bilirubin and ammonia, as demonstrated both in vitro using patient plasma and in vivo using ALF animal models. More importantly, we have also completed the 3-dimensional (3D) culturing of cells to meet the demands for industrialized rapid and mass production, and subsequently assembled the plasma-cell contacting BAL (PCC-BAL) system to fulfill the requirements of preclinical experiments. Extracorporeal blood purification of ALF rabbits with SynHeps-II-embedded PCC-BAL saved more than 80% of the animals from rapid death. Mechanistically, SynHeps-II therapy ameliorated liver and brain inflammation caused by high levels of bilirubin and ammonia and promoted liver regeneration by modulating the nuclear factor κB (NF-κB) and signal transducer and activator of transcription 3 (STAT3) pathways. Also, SynHeps-II treatment reduced cerebral infiltration of neutrophils, reduced reactive oxygen species (ROS) levels, and mitigated hepatic encephalopathy. Taken together, SynHeps-II cell-based BAL was promising for the treatment of ALF patients and warrants clinical trials.
Collapse
Affiliation(s)
- Ke Wang
- College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yuankui Zhu
- College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Mengqing Li
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yaxi Yang
- College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Dianbao Zuo
- College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Junfeng Sheng
- College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Xinhai Zhang
- College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Wei Wang
- Wuhan TOGO Medical Technology Co. Ltd., Wuhan, Hubei 430205, China
| | - Ping Zhou
- Wuhan TOGO Medical Technology Co. Ltd., Wuhan, Hubei 430205, China
| | - Mingqian Feng
- College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| |
Collapse
|
29
|
Liu H, Xie X, Wang Y, Wang X, Jin X, Zhang X, Wang Y, Zhu Z, Qi W, Jiang H. Development and validation of risk prediction model for bacterial infections in acute liver failure patients. Eur J Gastroenterol Hepatol 2024; 36:916-923. [PMID: 38829944 PMCID: PMC11136268 DOI: 10.1097/meg.0000000000002772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 03/15/2024] [Indexed: 06/05/2024]
Abstract
Infections significantly increase mortality in acute liver failure (ALF) patients, and there are no risk prediction models for early diagnosis and treatment of infections in ALF patients. This study aims to develop a risk prediction model for bacterial infections in ALF patients to guide rational antibiotic therapy. The data of ALF patients admitted to the Second Hospital of Hebei Medical University in China from January 2017 to January 2022 were retrospectively analyzed for training and internal validation. Patients were selected according to the updated 2011 American Association for the Study of Liver Diseases position paper on ALF. Serological indicators and model scores were collected within 24 h of admission. New models were developed using the multivariate logistic regression analysis. An optimal model was selected by receiver operating characteristic (ROC) analysis, Hosmer-Lemeshow test, the calibration curve, the Brier score, the bootstrap resampling, and the decision curve analysis. A nomogram was plotted to visualize the results. A total of 125 ALF patients were evaluated and 79 were included in the training set. The neutrophil-to-lymphocyte ratio and sequential organ failure assessment (SOFA) were integrated into the new model as independent predictive factors. The new SOFA-based model outperformed other models with an area under the ROC curve of 0.799 [95% confidence interval (CI): 0.652-0.926], the superior calibration and predictive performance in internal validation. High-risk individuals with a nomogram score ≥26 are recommended for antibiotic therapy. The new SOFA-based model demonstrates high accuracy and clinical utility in guiding antibiotic therapy in ALF patients.
Collapse
Affiliation(s)
- Huimin Liu
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases
| | - Xiaoli Xie
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases
| | - Yan Wang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases
| | - Xiaoting Wang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases
| | - Xiaoxu Jin
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases
| | - Xiaolin Zhang
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Hebei Province Key Laboratory of Environment and Human Health, Shijiazhuang, Hebei, China
| | - Yameng Wang
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Hebei Province Key Laboratory of Environment and Human Health, Shijiazhuang, Hebei, China
| | - Zongyi Zhu
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases
| | - Wei Qi
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases
| | - Huiqing Jiang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases
| |
Collapse
|
30
|
Tan N, Jian G, Peng J, Tian X, Chen B. Chishao - Fuzi herbal pair restore the macrophage M1/M2 balance in acute-on-chronic liver failure. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:118010. [PMID: 38499260 DOI: 10.1016/j.jep.2024.118010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 02/21/2024] [Accepted: 03/03/2024] [Indexed: 03/20/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional herbal pair Paeoniae Radix Rubra (roots of Paeonia lactiflora Pall., Chishao in Chinese) and Aconiti Lateralis Radix Praeparata (lateral roots of Aconitum carmichaelii Debeaux, Fuzi in Chinese) are widely used for the treatment of liver diseases, demonstrating clinical efficacy against acute-on-chronic liver failure (ACLF). As the core drug pair representing the "clearing method" and "warming method" in traditional Chinese medicine (TCM), they align with the TCM syndromic characteristics of ACLF, characterized by a mixture of deficiencies and realities. However, the molecular mechanisms underlying the anti-ACLF effects of Chishao - Fuzi herbal pair remain unclear. AIM OF THE STUDY To reveal the immunoinflammatory status of patients with hepatitis B virus-related ACLF (HBV-ACLF) based on macrophage polarization and to explore the mechanism of action of Chishao - Fuzi herbal pair in regulating macrophage polarization against ACLF. MATERIALS AND METHODS Peripheral blood samples were prospectively obtained from patients with HBV-ACLF, patients with chronic hepatitis B (CHB) in the immunoactive phase and healthy individuals. Flow cytometry, qRT-qPCR, and ELISA were used to reveal the activation status of monocyte-macrophages and the expression differences in related cytokines in the peripheral blood of patients with HBV-ACLF. Then, an ACLF rat model and a macrophage inflammation model in vitro were established. Hematoxylin-eosin staining, immunohistochemical staining, transmission electron microscopy, flow cytometry, western blotting, RT-qPCR, and ELISA were used to observe changes in the expression of M1/M2 macrophage markers and related inflammatory factors after Chishao - Fuzi herbal pair intervention, both in vivo and in vitro. RESULTS Patients with HBV-ACLF exhibited an imbalance in M1/M2 macrophage polarization, showing a tendency to activate M1 macrophages with high expression of CD86 and iNOS. This imbalance led to an increase in relevant pro-inflammatory factors (IL-1β, IL-6, TNF-α) and a decrease in anti-inflammatory factors (IL-10, TGF-β, VEGF), exacerbating the uncontrolled immune-inflammatory response. Chishao - Fuzi herbal pair intervention improved liver function, coagulation function, and histopathological injury in ACLF rats. It also partially ameliorated endotoxemia and inflammatory injury in ACLF. The mechanism was to restore the immune-inflammatory imbalance and prevent the exacerbation of inflammatory response to liver failure by promoting macrophage polarization toward M2 anti-inflammatory direction, inhibiting M1 macrophage activation, and increasing the levels of anti-inflammatory factors and decreasing pro-inflammatory factors. CONCLUSION Chishao - Fuzi herbal pair can reduce the systemic inflammatory burden of liver failure by modulating macrophage polarization and restoring ACLF immune-inflammatory imbalance. This study provides new perspectives and strategies for studying HBV-ACLF immune reconstitution and inflammatory response control.
Collapse
Affiliation(s)
- Nianhua Tan
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, China; Department of Hepatology, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410007, Hunan Province, China.
| | - Gonghui Jian
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, China
| | - Jie Peng
- Department of Hepatology, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410007, Hunan Province, China
| | - Xuefei Tian
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, China; Hunan Province University Key Laboratory of Oncology of Traditional Chinese Medicine Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China; Key Laboratory of Traditional Chinese Medicine for Mechanism of Tumor Prevention &Treatment, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
| | - Bin Chen
- Department of Hepatology, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410007, Hunan Province, China.
| |
Collapse
|
31
|
Tao Y, Wang Y, Wang M, Tang H, Chen E. Mesenchymal Stem Cells Alleviate Acute Liver Failure through Regulating Hepatocyte Apoptosis and Macrophage Polarization. J Clin Transl Hepatol 2024; 12:571-580. [PMID: 38974955 PMCID: PMC11224903 DOI: 10.14218/jcth.2023.00557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/06/2024] [Accepted: 03/18/2024] [Indexed: 07/09/2024] Open
Abstract
BACKGROUND AND AIMS Acute liver failure (ALF) is a life-threatening clinical problem with limited treatment options. Administration of human umbilical cord mesenchymal stem cells (hUC-MSCs) may be a promising approach for ALF. This study aimed to explore the role of hUC-MSCs in the treatment of ALF and the underlying mechanisms. METHODS A mouse model of ALF was induced by lipopolysaccharide and d-galactosamine administration. The therapeutic effects of hUC-MSCs were evaluated by assessing serum enzyme activity, histological appearance, and cell apoptosis in liver tissues. The apoptosis rate was analyzed in AML12 cells. The levels of inflammatory cytokines and the phenotype of RAW264.7 cells co-cultured with hUC-MSCs were detected. The C-Jun N-terminal kinase/nuclear factor-kappa B signaling pathway was studied. RESULTS The hUC-MSCs treatment decreased the levels of serum alanine aminotransferase and aspartate aminotransferase, reduced pathological damage, alleviated hepatocyte apoptosis, and reduced mortality in vivo. The hUC-MSCs co-culture reduced the apoptosis rate of AML12 cells in vitro. Moreover, lipopolysaccharide-stimulated RAW264.7 cells had higher levels of tumor necrosis factor-α, interleukin-6, and interleukin-1β and showed more CD86-positive cells, whereas the hUC-MSCs co-culture reduced the levels of the three inflammatory cytokines and increased the ratio of CD206-positive cells. The hUC-MSCs treatment inhibited the activation of phosphorylated (p)-C-Jun N-terminal kinase and p-nuclear factor-kappa B not only in liver tissues but also in AML12 and RAW264.7 cells co-cultured with hUC-MSCs. CONCLUSIONS hUC-MSCs could alleviate ALF by regulating hepatocyte apoptosis and macrophage polarization, thus hUC-MSC-based cell therapy may be an alternative option for patients with ALF.
Collapse
Affiliation(s)
- Yachao Tao
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Yonghong Wang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Menglan Wang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Enqiang Chen
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
32
|
Hu X, Li B, Lu B, Yu H, Du Y, Chen J. Identification and functional analysis of perforin 1 from largemouth bass (Micropterus salmoides). FISH & SHELLFISH IMMUNOLOGY 2024; 149:109531. [PMID: 38604479 DOI: 10.1016/j.fsi.2024.109531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 03/24/2024] [Accepted: 03/28/2024] [Indexed: 04/13/2024]
Abstract
In this study, we present the first cloning and identification of perforin (MsPRF1) in largemouth bass (Micropterus salmoides). The full-length cDNA of MsPRF1 spans 1572 base pairs, encoding a 58.88 kDa protein consisting of 523 amino acids. Notably, the protein contains MACPF and C2 structural domains. To evaluate the expression levels of MsPRF1 in various healthy largemouth bass tissues, real-time quantitative PCR was employed, revealing the highest expression in the liver and gut. After the largemouth bass were infected by Nocardia seriolae, the mRNA levels of MsPRF1 generally increased within 48 h. Remarkably, the recombinant protein MsPRF1 exhibits inhibitory effects against both Gram-negative and Gram-positive bacteria. Additionally, the largemouth bass showed a higher survival rate in the N. seriolae challenge following the intraperitoneal injection of rMsPRF1, with observed reductions in the tissue bacterial loads. Moreover, rMsPRF1 demonstrated a significant impact on the phagocytic and bactericidal activities of largemouth bass MO/MΦ cells, concurrently upregulating the expression of pro-inflammatory factors. These results demonstrate that MsPRF1 has a potential role in the immune response of largemouth bass against N. seriolae infection.
Collapse
Affiliation(s)
- Xiaoman Hu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Bin Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Bowen Lu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Hui Yu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Yang Du
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China.
| | - Jiong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
33
|
Brunese MC, Avella P, Cappuccio M, Spiezia S, Pacella G, Bianco P, Greco S, Ricciardelli L, Lucarelli NM, Caiazzo C, Vallone G. Future Perspectives on Radiomics in Acute Liver Injury and Liver Trauma. J Pers Med 2024; 14:572. [PMID: 38929793 PMCID: PMC11204538 DOI: 10.3390/jpm14060572] [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: 04/02/2024] [Revised: 05/02/2024] [Accepted: 05/09/2024] [Indexed: 06/28/2024] Open
Abstract
Background: Acute liver injury occurs most frequently due to trauma, but it can also occur because of sepsis or drug-induced injury. This review aims to analyze artificial intelligence (AI)'s ability to detect and quantify liver injured areas in adults and pediatric patients. Methods: A literature analysis was performed on the PubMed Dataset. We selected original articles published from 2018 to 2023 and cohorts with ≥10 adults or pediatric patients. Results: Six studies counting 564 patients were collected, including 170 (30%) children and 394 adults. Four (66%) articles reported AI application after liver trauma, one (17%) after sepsis, and one (17%) due to chemotherapy. In five (83%) studies, Computed Tomography was performed, while in one (17%), FAST-UltraSound was performed. The studies reported a high diagnostic performance; in particular, three studies reported a specificity rate > 80%. Conclusions: Radiomics models seem reliable and applicable to clinical practice in patients affected by acute liver injury. Further studies are required to achieve larger validation cohorts.
Collapse
Affiliation(s)
- Maria Chiara Brunese
- Department of Medicine and Health Science “V. Tiberio”, University of Molise, 86100 Campobasso, Italy; (M.C.B.)
| | - Pasquale Avella
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
- Hepatobiliary and Pancreatic Surgery Unit, Pineta Grande Hospital, 81030 Castel Volturno, Italy
| | - Micaela Cappuccio
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Salvatore Spiezia
- Department of Medicine and Health Science “V. Tiberio”, University of Molise, 86100 Campobasso, Italy; (M.C.B.)
| | - Giulia Pacella
- Department of Medicine and Health Science “V. Tiberio”, University of Molise, 86100 Campobasso, Italy; (M.C.B.)
| | - Paolo Bianco
- Hepatobiliary and Pancreatic Surgery Unit, Pineta Grande Hospital, 81030 Castel Volturno, Italy
| | - Sara Greco
- Interdisciplinary Department of Medicine, Section of Radiology and Radiation Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | | | - Nicola Maria Lucarelli
- Interdisciplinary Department of Medicine, Section of Radiology and Radiation Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Corrado Caiazzo
- Department of Medicine and Health Science “V. Tiberio”, University of Molise, 86100 Campobasso, Italy; (M.C.B.)
| | - Gianfranco Vallone
- Department of Medicine and Health Science “V. Tiberio”, University of Molise, 86100 Campobasso, Italy; (M.C.B.)
| |
Collapse
|
34
|
Shi H, Xie X, Zheng S, Chen H, Liu C, Li S, Lu M. Endotoxin tolerance ameliorates lipopolysaccharide/D-galactosamine-induced acute liver failure by negative regulation of the NF-κB/NLRP3 and activation of Nrf2/HO-1 via Sitr1. Int Immunopharmacol 2024; 132:111994. [PMID: 38581992 DOI: 10.1016/j.intimp.2024.111994] [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: 12/26/2023] [Revised: 03/26/2024] [Accepted: 03/30/2024] [Indexed: 04/08/2024]
Abstract
Acute liver failure (ALF) is a potentially fatal disorder characterized by extensive hepatocyte necrosis and rapid decline in liver function. Numerous factors, including oxidative stress, cell death, and inflammatory responses, are associated with its pathogenesis. Endotoxin tolerance (ET) refers to the phenomenon in which the body or cells exhibit low or no response to high-dose lipopolysaccharide (LPS) stimulation after pre-stimulation with low-dose LPS. However, the specific mechanism through which ET regulates LPS/D-galactosamine (D-GalN)-induced ALF remains unclear. An ALF mouse model was established by intraperitoneal injection of D-GalN (400 mg/kg) and LPS (10 mg/kg). A low dose of LPS (0.1 mg/kg/d) was continuously administered to mice for 5 d before modeling to assess the protective effect of ET. The data from this study showed that ET alleviated the inflammatory response in mice with LPS/D-GalN-induced ALF. ET inhibited LPS-induced oxidative damage and pyroptosis in macrophages in vitro. RNA sequencing analysis showed that the NF-κB/NLRP3 pathway was linked to the anti-inflammatory and antioxidative effects of ET. Furthermore, using western blot, RT-qPCR, and immunofluorescence, we verified that ET inhibited the NF-κB/NLRP3 pathway and triggered the Nrf2/HO-1 signaling pathway to attenuate oxidative stress and cell pyroptosis. Sirt1 knockdown reversed this protective effect. In summary, our research elucidates that ET prevents ALF advancement by upregulating Sirt1 levels, triggering the Nrf2/HO-1 signaling axis, and suppressing the NF-κB/NLRP3 signaling cascade to inhibit oxidative stress and cell pyroptosis. Our results provide a mechanistic explanation for the protective effect of ET against ALF.
Collapse
Affiliation(s)
- Huifang Shi
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xueting Xie
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Sijie Zheng
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hong Chen
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chenyi Liu
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shu Li
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Mingqin Lu
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
35
|
Artru F, McPhail MJ. Immunopathogenesis of acute on chronic liver failure. Am J Transplant 2024; 24:724-732. [PMID: 38346497 DOI: 10.1016/j.ajt.2024.02.001] [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: 09/30/2023] [Revised: 01/13/2024] [Accepted: 02/01/2024] [Indexed: 02/23/2024]
Abstract
Acute-on-chronic liver failure is a well-established description of a high-mortality syndrome of chronic liver disease (usually cirrhosis) with organ failure. While the exact definition is under refinement, the accepted understanding of this entity is in patients with chronic liver disease and various organs in failure and where systemic inflammation is a major component of the pathobiology. There are limited therapies for a disease with such a poor prognosis, and while improvements in the critical care management and for very few patients, liver transplantation, mean 50% can survive to hospital discharge, rapid application of new therapies is required. Here we explain the current understanding of the immunologic abnormalities seen in acute-on-chronic liver failure across the innate and adaptive immune systems, the role of the hepatic cell death and the gut-liver axis, and recommendations for future research and treatment paradigms.
Collapse
Affiliation(s)
- Florent Artru
- Institute of Liver Studies, King's College Hospital, London, United Kingdom; Department of Inflammation Biology, School of Immunology and Microbial Sciences, King's College London, United Kingdom; Liver department and NUMECAN institute, Rennes University Hospital and Rennes University, France
| | - Mark J McPhail
- Institute of Liver Studies, King's College Hospital, London, United Kingdom; Department of Inflammation Biology, School of Immunology and Microbial Sciences, King's College London, United Kingdom.
| |
Collapse
|
36
|
Umbaugh DS, Nguyen NT, Smith SH, Ramachandran A, Jaeschke H. The p21 + perinecrotic hepatocytes produce the chemokine CXCL14 after a severe acetaminophen overdose promoting hepatocyte injury and delaying regeneration. Toxicology 2024; 504:153804. [PMID: 38614205 PMCID: PMC11108579 DOI: 10.1016/j.tox.2024.153804] [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: 02/08/2024] [Revised: 03/30/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
Fifty percent of all acute liver failure (ALF) cases in the United States are due to acetaminophen (APAP) overdose. Assessment of canonical features of liver injury, such as plasma alanine aminotransferase activities are poor predictors of acute liver failure (ALF), suggesting the involvement of additional mechanisms independent of hepatocyte death. Previous work demonstrated a severe overdose of APAP results in impaired regeneration, the induction of senescence by p21, and increased mortality. We hypothesized that a discrete population of p21+ hepatocytes acquired a secretory phenotype that directly impedes liver recovery after a severe APAP overdose. Leveraging in-house human APAP explant liver and publicly available single-nuclei RNAseq data, we identified a subpopulation of p21+ hepatocytes enriched in a unique secretome of factors, such as CXCL14. Spatial transcriptomics in the mouse model of APAP overdose confirmed the presence of a p21+ hepatocyte population that directly surrounded the necrotic areas. In both male and female mice, we found a dose-dependent induction of p21 and persistent circulating levels of the p21-specific constituent, CXCL14, in the plasma after a severe APAP overdose. In parallel experiments, we targeted either the putative senescent hepatocytes with the senolytic drugs, dasatinib and quercetin, or CXCL14 with a neutralizing antibody. We found that targeting CXCL14 greatly enhanced liver recovery after APAP-induced liver injury, while targeting senescent hepatocytes had no effect. These data support the conclusion that the sustained induction of p21 in hepatocytes with persistent CXCL14 secretion are critical mechanistic events leading to ALF in mice and human patients.
Collapse
Affiliation(s)
- David S Umbaugh
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Nga T Nguyen
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Sawyer H Smith
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Anup Ramachandran
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
37
|
Liu L, Lin L, Wang Y, Yan X, Li R, He M, Li H, Zhuo C, Li L, Zhang D, Wang X, Huang W, Li X, Mao Y, Chen H, Wu S, Jiang W, Zhu L. L-AP Alleviates Liver Injury in Septic Mice by Inhibiting Macrophage Activation via Suppressing NF-κB and NLRP3 Inflammasome/Caspase-1 Signal Pathways. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:8460-8475. [PMID: 38564364 DOI: 10.1021/acs.jafc.3c02781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Liver injury and progressive liver failure are severe life-threatening complications in sepsis, further worsening the disease and leading to death. Macrophages and their mediated inflammatory cytokine storm are critical regulators in the occurrence and progression of liver injury in sepsis, for which effective treatments are still lacking. l-Ascorbic acid 6-palmitate (L-AP), a food additive, can inhibit neuroinflammation by modulating the phenotype of the microglia, but its pharmacological action in septic liver damage has not been fully explored. We aimed to investigate L-AP's antisepticemia action and the possible pharmacological mechanisms in attenuating septic liver damage by modulating macrophage function. We observed that L-AP treatment significantly increased survival in cecal ligation and puncture-induced WT mice and attenuated hepatic inflammatory injury, including the histopathology of the liver tissues, hepatocyte apoptosis, and the liver enzyme levels in plasma, which were comparable to NLRP3-deficiency in septic mice. L-AP supplementation significantly attenuated the excessive inflammatory response in hepatic tissues of septic mice in vivo and in cultured macrophages challenged by both LPS and ATP in vitro, by reducing the levels of NLRP3, pro-IL-1β, and pro-IL-18 mRNA expression, as well as the levels of proteins for p-I-κB-α, p-NF-κB-p65, NLRP3, cleaved-caspase-1, IL-1β, and IL-18. Additionally, it impaired the inflammasome ASC spot activation and reduced the inflammatory factor contents, including IL-1β and IL-18 in plasma/cultured superannuants. It also prevented the infiltration/migration of macrophages and their M1-like inflammatory polarization while improving their M2-like polarization. Overall, our findings revealed that L-AP protected against sepsis by reducing macrophage activation and inflammatory cytokine production by suppressing their activation in NF-κB and NLRP3 inflammasome signal pathways in septic liver.
Collapse
Affiliation(s)
- Linling Liu
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Lan Lin
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Yingling Wang
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xin Yan
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Ruli Li
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Min He
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - He Li
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Caili Zhuo
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Lingyu Li
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Die Zhang
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xuemei Wang
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Wenjing Huang
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xinyue Li
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Yan Mao
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Hongying Chen
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Sisi Wu
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Wei Jiang
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Ling Zhu
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| |
Collapse
|
38
|
Sauer J, Steixner-Kumar AA, Gabler S, Motyka M, Rippmann JF, Brosa S, Boettner D, Schönberger T, Lempp C, Frodermann V, Simon E, Krenkel O, Bahrami E. Diverse potential of secretome from natural killer cells and monocyte-derived macrophages in activating stellate cells. Front Immunol 2024; 15:1232070. [PMID: 38638443 PMCID: PMC11025356 DOI: 10.3389/fimmu.2024.1232070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 03/04/2024] [Indexed: 04/20/2024] Open
Abstract
Chronic liver diseases, such as non-alcoholic steatohepatitis (NASH)-induced cirrhosis, are characterized by an increasing accumulation of stressed, damaged, or dying hepatocytes. Hepatocyte damage triggers the activation of resident immune cells, such as Kupffer cells (KC), as well as the recruitment of immune cells from the circulation toward areas of inflammation. After infiltration, monocytes differentiate into monocyte-derived macrophages (MoMF) which are functionally distinct from resident KC. We herein aim to compare the in vitro signatures of polarized macrophages and activated hepatic stellate cells (HSC) with ex vivo-derived disease signatures from human NASH. Furthermore, to shed more light on HSC activation and liver fibrosis progression, we investigate the effects of the secretome from primary human monocytes, macrophages, and NK cells on HSC activation. Interleukin (IL)-4 and IL-13 treatment induced transforming growth factor beta 1 (TGF-β1) secretion by macrophages. However, the supernatant transfer did not induce HSC activation. Interestingly, PMA-activated macrophages showed strong induction of the fibrosis response genes COL10A1 and CTGF, while the supernatant of IL-4/IL-13-treated monocytes induced the upregulation of COL3A1 in HSC. The supernatant of PMA-activated NK cells had the strongest effect on COL10A1 induction in HSC, while IL-15-stimulated NK cells reduced the expression of COL1A1 and CTGF. These data indicate that other factors, aside from the well-known cytokines and chemokines, might potentially be stronger contributors to the activation of HSCs and induction of a fibrotic response, indicating a more diverse and complex role of monocytes, macrophages, and NK cells in liver fibrosis progression.
Collapse
Affiliation(s)
- Julia Sauer
- Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | - Svenja Gabler
- Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | | | - Stefan Brosa
- Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Dennis Boettner
- Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | - Charlotte Lempp
- Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | - Eric Simon
- Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Oliver Krenkel
- Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Ehsan Bahrami
- Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| |
Collapse
|
39
|
Feng L, Wang Y, Fu Y, Li T, He G. Stem Cell-Based Strategies: The Future Direction of Bioartificial Liver Development. Stem Cell Rev Rep 2024; 20:601-616. [PMID: 38170319 DOI: 10.1007/s12015-023-10672-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2023] [Indexed: 01/05/2024]
Abstract
Acute liver failure (ALF) results from severe liver damage or end-stage liver disease. It is extremely fatal and causes serious health and economic burdens worldwide. Once ALF occurs, liver transplantation (LT) is the only definitive and recommended treatment; however, LT is limited by the scarcity of liver grafts. Consequently, the clinical use of bioartificial liver (BAL) has been proposed as a treatment strategy for ALF. Human primary hepatocytes are an ideal cell source for these methods. However, their high demand and superior viability prevent their widespread use. Hence, finding alternatives that meet the seed cell quality and quantity requirements is imperative. Stem cells with self-renewing, immunogenic, and differentiative capacities are potential cell sources. MSCs and its secretomes encompass a spectrum of beneficial properties, such as anti-inflammatory, immunomodulatory, anti-ROS (reactive oxygen species), anti-apoptotic, pro-metabolomic, anti-fibrogenesis, and pro-regenerative attributes. This review focused on the recent status and future directions of stem cell-based strategies in BAL for ALF. Additionally, we discussed the opportunities and challenges associated with promoting such strategies for clinical applications.
Collapse
Affiliation(s)
- Lei Feng
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China.
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550000, Guizhou, China.
| | - Yi Wang
- Shanxi Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, Shanxi, China
| | - Yu Fu
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Ting Li
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China.
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510140, Guangdong, China.
| | - Guolin He
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China.
| |
Collapse
|
40
|
Xu M, Chen Y, Artru F. Acute decompensation of cirrhosis versus acute-on-chronic liver failure: What are the clinical implications? United European Gastroenterol J 2024; 12:194-202. [PMID: 38376886 PMCID: PMC10954432 DOI: 10.1002/ueg2.12538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 12/20/2023] [Indexed: 02/21/2024] Open
Abstract
It is essential to identify the subgroup of patients who experience poorer outcomes in order to adapt clinical management effectively. In the context of liver disease, the earlier the identification occurs, the greater the range of therapeutic options that can be offered to patients. In the past, patients with acute decompensation (AD) of chronic liver disease were treated as a homogeneous group, with emphasis on identifying those at the highest risk of death. In the last 15 years, a differentiation has emerged between acute-on-chronic liver failure syndrome (ACLF) and AD, primarily due to indications that the latter is linked to a less favorable short-term prognosis. Nevertheless, the definition of ACLF varies among the different knowledge societies, making it challenging to assess its true impact compared with AD. Therefore, the purpose of this review is to provide a detailed analysis emphasizing the critical importance of identifying ACLF in the field of advanced liver disease. We will discuss the differences between Eastern and Western approaches, particularly in relation to the occurrence of liver failure and disease onset. Common characteristics, such as the dynamic nature of the disease course, will be highlighted. Finally, we will focus on two key clinical implications arising from these considerations: the prevention of ACLF before its onset and the clinical management strategies once it develops, including liver transplantation and withdrawal of care.
Collapse
Affiliation(s)
- Manman Xu
- Fourth Department of Liver Disease (Difficult & Complicated Liver Diseases and Artificial Liver Center)Beijing You'an Hospital Affiliated to CapitalMedical UniversityBeijingChina
- Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment ResearchBeijingChina
| | - Yu Chen
- Fourth Department of Liver Disease (Difficult & Complicated Liver Diseases and Artificial Liver Center)Beijing You'an Hospital Affiliated to CapitalMedical UniversityBeijingChina
- Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment ResearchBeijingChina
| | - Florent Artru
- Institute of Liver StudiesKing's College HospitalLondonUK
- Liver Disease DepartmentRennes University HospitalRennesFrance
- Rennes University and Inserm NuMeCan UMR 1317RennesFrance
| |
Collapse
|
41
|
Liu J, Zhang Q, Wong YK, Luo P, Chen J, Xie L, Chen J, He X, Shi F, Gong P, Liu X, Wang J. Single-Cell Transcriptomics Reveals the Ameliorative Effect of Oridonin on Septic Liver Injury. Adv Biol (Weinh) 2024; 8:e2300542. [PMID: 38408269 DOI: 10.1002/adbi.202300542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/23/2023] [Indexed: 02/28/2024]
Abstract
Sepsis is a life-threatening syndrome leading to hemodynamic instability and potential organ dysfunction. Oridonin, commonly used in Traditional Chinese Medicine (TCM), exhibits significant anti-inflammation activity. To explore the protective mechanisms of oridonin against the pathophysiological changes, the authors conducted single-cell transcriptome (scRNA-seq) analysis on septic liver models induced by cecal ligation and puncture (CLP). They obtained a total of 63,486 cells, distributed across 11 major cell clusters, and concentrated their analysis on four specific clusters (hepatocytes/Heps, macrophages, endothelial/Endos and T/NK) based on their changes in proportion during sepsis and under oridonin treatment. Firstly, biological changes in Hep, which are related to metabolic dysregulation and pro-inflammatory signaling, are observed during sepsis. Secondly, they uncovered the dynamic profiles of macrophage's phenotype, indicating that a substantial number of macrophages exhibited a M1-skewed phenotype associated with pro-inflammatory characteristics in septic model. Thirdly, they detected an upregulation of both inflammatory cytokines and transcriptomic factor Nfkb1 expression within Endo, along with slight capillarization during sepsis. Moreover, excessive accumulation of cytotoxic NK led to an immune imbalance. Though, oridonin ameliorated inflammatory-related responses and improved the liver dysfunction in septic mice. This study provides fundamental evidence of the protective effects of oridonin against sepsis-induced cytokine storm.
Collapse
Affiliation(s)
- Jing Liu
- Department of Critical Medicine, Shenzhen Institute of Respiratory Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, Guangdong, 518020, China
| | - Qian Zhang
- School of Traditional Chinese Medicine and School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yin Kwan Wong
- Department of Critical Medicine, Shenzhen Institute of Respiratory Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, Guangdong, 518020, China
| | - Piao Luo
- School of Traditional Chinese Medicine and School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Junhui Chen
- Department of Critical Medicine, Shenzhen Institute of Respiratory Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, Guangdong, 518020, China
| | - Lulin Xie
- Department of Critical Medicine, Shenzhen Institute of Respiratory Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, Guangdong, 518020, China
| | - Jiayun Chen
- School of Traditional Chinese Medicine and School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Xueling He
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Fei Shi
- Department of Infectious Disease, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, China
| | - Ping Gong
- Department of Emergency, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, China
| | - Xueyan Liu
- Department of Critical Medicine, Shenzhen Institute of Respiratory Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, Guangdong, 518020, China
| | - Jigang Wang
- Department of Critical Medicine, Shenzhen Institute of Respiratory Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, Guangdong, 518020, China
- School of Traditional Chinese Medicine and School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
| |
Collapse
|
42
|
Yang Y, Tian T, Wang Z, Li S, Li N, Luo H, Jiang Y. LncRNA 220, a newly discovered long non-conding RNA inhibiting apoptosis and autophagy in Kupffer cells in LPS-induced endotoxemic mice through the XBP1u-PI3K-AKT pathway. Int Immunopharmacol 2024; 128:111497. [PMID: 38241842 DOI: 10.1016/j.intimp.2024.111497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/20/2023] [Accepted: 01/03/2024] [Indexed: 01/21/2024]
Abstract
Sepsis is recognized as a potentially fatal condition characterized by acute organ dysfunction resulting from an imbalanced immune response to infection. Acute liver injury (ALI) arises as an inflammatory outcome of immune response dysregulation associated with sepsis. Kupffer cells, which are liver-specific macrophages, are known to have a significant impact on ALI, although the precise regulatory mechanism remains unclear. Numerous studies have showcased the regulatory impact of long non-coding RNAs (lncRNAs) on the progression of diverse ailments, yet their precise regulatory mechanisms remain predominantly unexplored. In this study, a novel long non-coding RNA (lncRNA), referred to as lncRNA 220, was discovered using high-throughput sequencing. The expression of lncRNA 220 was found to be significantly elevated in the livers of mice with lipopolysaccharide (LPS)-induced endotoxemia, specifically during the 8-hour time period. Furthermore, in Kupffer cells treated with LPS, lncRNA 220 was observed to inhibit apoptosis and autophagy by activating the PI3K-AKT-mTORC1 pathway. This effect was achieved through the reduction of X-box protein 1 unspliced (Xbp1u) mRNA stability and suppression of its translation in the context of endoplasmic reticulum stress (ERS). Ultimately, this intervention mitigated the progression of LPS-induced ALI. To summarize, our study establishes lncRNA 220 as a newly identified regulator that suppresses apoptosis and autophagy in Kupffer cells subjected to LPS treatment, indicating its potential as a molecular target for ALI in endotoxemic mice.
Collapse
Affiliation(s)
- Ying Yang
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Tian Tian
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhenqi Wang
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shan Li
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Nanhong Li
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Haihua Luo
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yong Jiang
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Department of Respiratory and Critical Care Medicine, Affiliated Dongguan Hospital, Southern Medical University, Guangdong, China.
| |
Collapse
|
43
|
Ni L, Chen D, Zhao Y, Ye R, Fang P. Unveiling the flames: macrophage pyroptosis and its crucial role in liver diseases. Front Immunol 2024; 15:1338125. [PMID: 38380334 PMCID: PMC10877142 DOI: 10.3389/fimmu.2024.1338125] [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: 11/14/2023] [Accepted: 01/16/2024] [Indexed: 02/22/2024] Open
Abstract
Macrophages play a critical role in innate immunity, with approximately 90% of the total macrophage population in the human body residing in the liver. This population encompasses both resident and infiltrating macrophages. Recent studies highlight the pivotal role of liver macrophages in various aspects such as liver inflammation, regeneration, and immune regulation. A novel pro-inflammatory programmed cell death, pyroptosis, initially identified in macrophages, has garnered substantial attention since its discovery. Studies investigating pyroptosis and inflammation progression have particularly centered around macrophages. In liver diseases, pyroptosis plays an important role in driving the inflammatory response, facilitating the fibrotic process, and promoting tumor progression. Notably, the role of macrophage pyroptosis cannot be understated. This review primarily focuses on the role of macrophage pyroptosis in liver diseases. Additionally, it underscores the therapeutic potential inherent in targeting macrophage pyroptosis.
Collapse
Affiliation(s)
| | | | | | | | - Peng Fang
- Department of Infectious Diseases, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| |
Collapse
|
44
|
Cui A, Li B, Wallace MS, Gonye ALK, Oetheimer C, Patel H, Tonnerre P, Holmes JA, Lieb D, Yao BS, Ma A, Roberts K, Damasio M, Chen JH, Piou D, Carlton-Smith C, Brown J, Mylvaganam R, Hon Fung JM, Sade-Feldman M, Aneja J, Gustafson J, Epstein ET, Salloum S, Brisac C, Thabet A, Kim AY, Lauer GM, Hacohen N, Chung RT, Alatrakchi N. Single-cell atlas of the liver myeloid compartment before and after cure of chronic viral hepatitis. J Hepatol 2024; 80:251-267. [PMID: 36972796 PMCID: PMC11651724 DOI: 10.1016/j.jhep.2023.02.040] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 01/22/2023] [Accepted: 02/14/2023] [Indexed: 03/29/2023]
Abstract
BACKGROUND & AIMS Chronic viral infections present serious public health challenges; however, direct-acting antivirals (DAAs) are now able to cure nearly all patients infected with hepatitis C virus (HCV), representing the only cure of a human chronic viral infection to date. DAAs provide a valuable opportunity to study immune pathways in the reversal of chronic immune failures in an in vivo human system. METHODS To leverage this opportunity, we used plate-based single-cell RNA-seq to deeply profile myeloid cells from liver fine needle aspirates in patients with HCV before and after DAA treatment. We comprehensively characterised liver neutrophils, eosinophils, mast cells, conventional dendritic cells, plasmacytoid dendritic cells, classical monocytes, non-classical monocytes, and macrophages, and defined fine-grained subpopulations of several cell types. RESULTS We discovered cell type-specific changes post-cure, including an increase in MCM7+STMN1+ proliferating CD1C+ conventional dendritic cells, which may support restoration from chronic exhaustion. We observed an expected downregulation of interferon-stimulated genes (ISGs) post-cure as well as an unexpected inverse relationship between pre-treatment viral load and post-cure ISG expression in each cell type, revealing a link between viral loads and sustained modifications of the host's immune system. We found an upregulation of PD-L1/L2 gene expression in ISG-high neutrophils and IDO1 expression in eosinophils, pinpointing cell subpopulations crucial for immune regulation. We identified three recurring gene programmes shared by multiple cell types, distilling core functions of the myeloid compartment. CONCLUSIONS This comprehensive single-cell RNA-seq atlas of human liver myeloid cells in response to cure of chronic viral infections reveals principles of liver immunity and provides immunotherapeutic insights. CLINICAL TRIAL REGISTRATION This study is registered at ClinicalTrials.gov (NCT02476617). IMPACT AND IMPLICATIONS Chronic viral liver infections continue to be a major public health problem. Single-cell characterisation of liver immune cells during hepatitis C and post-cure provides unique insights into the architecture of liver immunity contributing to the resolution of the first curable chronic viral infection of humans. Multiple layers of innate immune regulation during chronic infections and persistent immune modifications after cure are revealed. Researchers and clinicians may leverage these findings to develop methods to optimise the post-cure environment for HCV and develop novel therapeutic approaches for other chronic viral infections.
Collapse
Affiliation(s)
- Ang Cui
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Bo Li
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Harvard University Virology Program, Harvard Medical School, Boston, MA, USA
| | - Michael S Wallace
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Anna L K Gonye
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Christopher Oetheimer
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Hailey Patel
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Pierre Tonnerre
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Institut de Recherche Saint-Louis, Université Paris Cité, Inserm U976 (HIPI), Team ATIP-Avenir, Paris, France
| | - Jacinta A Holmes
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Department of Gastroenterology, St Vincent's Hospital Melbourne, Melbourne, VIC, Australia
| | - David Lieb
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Brianna S Yao
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Aileen Ma
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kela Roberts
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Marcos Damasio
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jonathan H Chen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA; Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Daphnee Piou
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Charles Carlton-Smith
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Joelle Brown
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ravi Mylvaganam
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | | | - Moshe Sade-Feldman
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Jasneet Aneja
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Division of Infectious Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jenna Gustafson
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Eliana T Epstein
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Shadi Salloum
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Cynthia Brisac
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ashraf Thabet
- Department of Interventional Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Arthur Y Kim
- Division of Infectious Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Georg M Lauer
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Nir Hacohen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA; Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Raymond T Chung
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Nadia Alatrakchi
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
45
|
Li J, Huang Q, Ma W, Yi J, Zhong X, Hu R, Sun J, Ma M, Lv M, Han Z, Zhang W, Feng W, Sun X, Zhou X. Hepatoprotective efficacy and interventional mechanism of JianPi LiShi YangGan formula in acute-on-chronic liver failure. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116880. [PMID: 37422102 DOI: 10.1016/j.jep.2023.116880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/24/2023] [Accepted: 07/05/2023] [Indexed: 07/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Acute-on-chronic liver failure (ACLF) progresses rapidly with a high short-term death rate. Although JianPi LiShi YangGan formula (YGF) has been used to treat ACLF by managing inflammatory responses and reducing endotoxemia, hepatocyte injury, and mortality, the underlying mechanisms remain unclear. AIM OF THE STUDY This study aims to investigate the potential mechanisms underlying the efficacy and protective benefits of YGF in mice with ACLF. MATERIALS AND METHODS YGF composition was determined using high-performance liquid chromatography coupled with mass spectrometry. We constructed a mouse model of ACLF using carbon tetrachloride, lipopolysaccharide (LPS), and D-galactosamine (D-Gal), as well as an in vitro model of D-Gal/LPS-induced hepatocyte injury. The therapeutic effects of YGF in ACLF mice were verified using hematoxylin-eosin, Sirius red, and Masson staining, and by measuring serum alanine transaminase (ALT), aspartate transaminase (AST), and inflammatory cytokine levels. Mitochondrial damage in hepatocytes was evaluated using electron microscopy, while superoxide anion levels in liver tissue were investigated using dihydroethidium. Transcriptome analysis, immunohistochemistry, western blotting, and immunofluorescence assays were performed to explore the mechanisms underlying the ameliorative effects of YGF against ACLF. RESULTS In mice with ACLF, YGF therapy partially decreased serum inflammatory cytokine levels, as well as hepatocyte injury and liver fibrosis. The livers of ACLF mice treated with YGF exhibited decreased mitochondrial damage and reactive oxygen species generation, as well as a decreased number of M1 macrophages and increased number of M2 macrophages. Transcriptome analysis revealed that YGF may regulate biological processes such as autophagy, mitophagy, and PI3K/AKT signaling. In ACLF mice, YGF promoted mitophagy and inhibited PI3K/AKT/mTOR pathway activation in hepatocytes. Meanwhile, the autophagy inhibitor 3M-A reduced the capacity of YGF to induce autophagy and protect against hepatocyte injury in vitro. In contrast, the PI3K agonist 740 Y-P suppressed the ability of YGF to control PI3K/AKT/mTOR pathway activation and induce autophagy. CONCLUSIONS Together, our findings suggest that YGF mediates autophagy, tight junctions, cytokine generation, and other biological processes. In addition, YGF inhibits hepatic inflammatory responses and ameliorates hepatocyte injury in mice with ACLF. Mechanistically, YGF can promote mitophagy to ameliorate acute-on-chronic liver failure by inhibiting the PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Jing Li
- Macau University of Science and Technology, Faculty of Chinese Medicine, Taipa, Macau; Shenzhen Traditional Chinese Medicine Hospital, Department of Liver Disease, Shenzhen, 518033, China; The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Department of Liver Disease, Shenzhen, 518033, China
| | - Qi Huang
- Shenzhen Traditional Chinese Medicine Hospital, Department of Liver Disease, Shenzhen, 518033, China; The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Department of Liver Disease, Shenzhen, 518033, China
| | - Wenfeng Ma
- Shenzhen Traditional Chinese Medicine Hospital, Department of Liver Disease, Shenzhen, 518033, China; The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Department of Liver Disease, Shenzhen, 518033, China
| | - JinYu Yi
- Macau University of Science and Technology, Faculty of Chinese Medicine, Taipa, Macau; Shenzhen Traditional Chinese Medicine Hospital, Department of Liver Disease, Shenzhen, 518033, China; The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Department of Liver Disease, Shenzhen, 518033, China
| | - Xin Zhong
- Shenzhen Traditional Chinese Medicine Hospital, Department of Liver Disease, Shenzhen, 518033, China; The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Department of Liver Disease, Shenzhen, 518033, China
| | - Rui Hu
- Macau University of Science and Technology, Faculty of Chinese Medicine, Taipa, Macau; Shenzhen Traditional Chinese Medicine Hospital, Department of Liver Disease, Shenzhen, 518033, China; The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Department of Liver Disease, Shenzhen, 518033, China
| | - Jialing Sun
- Shenzhen Traditional Chinese Medicine Hospital, Department of Liver Disease, Shenzhen, 518033, China; The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Department of Liver Disease, Shenzhen, 518033, China
| | - MengQing Ma
- Shenzhen Traditional Chinese Medicine Hospital, Department of Liver Disease, Shenzhen, 518033, China; The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Department of Liver Disease, Shenzhen, 518033, China
| | - Minling Lv
- Shenzhen Traditional Chinese Medicine Hospital, Department of Liver Disease, Shenzhen, 518033, China; The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Department of Liver Disease, Shenzhen, 518033, China
| | - Zhiyi Han
- Shenzhen Traditional Chinese Medicine Hospital, Department of Liver Disease, Shenzhen, 518033, China; The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Department of Liver Disease, Shenzhen, 518033, China
| | - Wei Zhang
- Shenzhen Traditional Chinese Medicine Hospital, Department of Liver Disease, Shenzhen, 518033, China; The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Department of Liver Disease, Shenzhen, 518033, China
| | - Wenxing Feng
- Shenzhen Traditional Chinese Medicine Hospital, Department of Liver Disease, Shenzhen, 518033, China; The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Department of Liver Disease, Shenzhen, 518033, China
| | - Xinfeng Sun
- Shenzhen Traditional Chinese Medicine Hospital, Department of Liver Disease, Shenzhen, 518033, China; The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Department of Liver Disease, Shenzhen, 518033, China
| | - Xiaozhou Zhou
- Shenzhen Traditional Chinese Medicine Hospital, Department of Liver Disease, Shenzhen, 518033, China; The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Department of Liver Disease, Shenzhen, 518033, China.
| |
Collapse
|
46
|
Xuan R, Qiu X, Wang J, Liu S, Magnuson JT, Xu B, Qiu W, Zheng C. Hepatotoxic response of perfluorooctane sulfonamide (PFOSA) in early life stage zebrafish (Danio rerio) is greater than perfluorooctane sulfonate (PFOS). JOURNAL OF HAZARDOUS MATERIALS 2024; 461:132552. [PMID: 37716261 DOI: 10.1016/j.jhazmat.2023.132552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 09/12/2023] [Accepted: 09/12/2023] [Indexed: 09/18/2023]
Abstract
Perfluorooctane sulfonamide (PFOSA), a typical perfluorooctane sulfonate precursor (PreFOS), has been detected in the aquatic environment globally. However, the effects of PFOSA at levels measured in the environment have not been well characterized in aquatic organisms. In this study, we evaluated the transcriptional, biochemical, histopathological, and morphological effects of PFOSA to characterize the underlying mechanisms of toxicity by using a universal model in aquatic ecotoxicology, zebrafish (Danio rerio). Transcriptional changes in PFOSA-exposed zebrafish predicted hepatic fibrosis and associated immune function. Subsequent, sublethal impacts were observed, which included significant alterations in liver-specific protein levels, increased immune cell numbers, and liver pathological structural damage. In addition, we compared the effects caused by PFOSA and perfluorooctane sulfonate (PFOS) at the same exposure concentration and found a greater hepatotoxic effect of PFOSA relative to PFOS, indicating that the adverse impacts of PFOSA may be more severe. This was the first study to comparatively explore the hepatotoxic response of PFOSA and PFOS in aquatic organisms, which can be used for ecological risk assessments of PreFOS compounds.
Collapse
Affiliation(s)
- Rongrong Xuan
- The First Affiliated Hospital of Ningbo University, Ningbo 315020, China
| | - Xiaojian Qiu
- The First Affiliated Hospital of Ningbo University, Ningbo 315020, China
| | - Jiazhen Wang
- Guangdong Provincial Key Laboratory of Soil and Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, 1088 Xueyuan Avenue, Nanshan District, Shenzhen 518055, China
| | - Shuai Liu
- Institute of Microbiology, Jiangxi Academy of Sciences, Nanchang 330096, China
| | - Jason T Magnuson
- US Geological Survey, Columbia Environmental Research Center, Columbia, MO, USA
| | - Bentuo Xu
- National and Local Joint Engineering Research Center of Ecological Treatment Technology for Urban Water Pollution, School of Life and Environmental Science, Wenzhou University, Chashan University Town, Wenzhou 325035, China
| | - Wenhui Qiu
- The First Affiliated Hospital of Ningbo University, Ningbo 315020, China; Guangdong Provincial Key Laboratory of Soil and Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, 1088 Xueyuan Avenue, Nanshan District, Shenzhen 518055, China.
| | - Chunmiao Zheng
- Guangdong Provincial Key Laboratory of Soil and Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, 1088 Xueyuan Avenue, Nanshan District, Shenzhen 518055, China; EIT Institute for Advanced Study, 568 Tongxin Road, Zhenhai District, Ningbo 315410, China
| |
Collapse
|
47
|
Hirsch W, Megna B, Adeyi O, Lim N. Macrophage activation syndrome with acute hepatitis in a patient with adult-onset immunodeficiency with anti-interferon gamma antibodies: a case report. J Med Case Rep 2024; 18:6. [PMID: 38178160 PMCID: PMC10768153 DOI: 10.1186/s13256-023-04245-w] [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: 08/02/2023] [Accepted: 11/05/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Macrophage activation syndrome is a rare disorder leading to unregulated immune activity manifesting with nonspecific constitutional symptoms, laboratory abnormalities, and multiorgan involvement. We report the case of a patient who presented with acute hepatitis secondary to macrophage activation syndrome diagnosed by liver biopsy and successfully treated with intravenous immune globulin, anakinra, and rituximab. CASE PRESENTATION A 42-year-old Laotian woman with adult-onset immunodeficiency with anti-interferon gamma antibodies presented with a fever, headache, generalized myalgia, dark urine, and reduced appetite in the setting of family members at home with similar symptoms. Her laboratory workup was notable for evidence of acute hepatitis without acute liver failure. After an unrevealing comprehensive infectious and noninvasive rheumatologic workup was completed, a liver biopsy was performed ultimately revealing the diagnosis of macrophage activation syndrome. She was successfully treated with intravenous immune globulin, anakinra, and rituximab. CONCLUSION This case highlights the importance of maintaining macrophage activation syndrome on the differential of a patient with acute hepatitis of unknown etiology in the correct clinical context and the value of a liver biopsy in making a diagnosis when noninvasive testing is unrevealing.
Collapse
Affiliation(s)
- William Hirsch
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA.
| | - Bryant Megna
- Division of Gastroenterology, Hepatology, & Nutrition, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Oyedele Adeyi
- Department of Laboratory Medicine & Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Nicholas Lim
- Division of Gastroenterology, Hepatology, & Nutrition, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
48
|
Ye Q, Wang H, Chen Y, Zheng Y, Du Y, Ma C, Zhang Q. PANoptosis-like death in acute-on-chronic liver failure injury. Sci Rep 2024; 14:392. [PMID: 38172209 PMCID: PMC10764922 DOI: 10.1038/s41598-023-50720-1] [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: 04/01/2023] [Accepted: 12/23/2023] [Indexed: 01/05/2024] Open
Abstract
The pathogenesis of Acute-on-chronic liver failure (ACLF) involves several forms of cell death, such as pyroptosis, apoptosis, and necroptosis, which consist of PANoptosis. To explore PANoptosis as a regulated cell death pathway in ACLF. Firstly, a bioinformatic strategy was used to observe the role of the PANoptosis pathway in ACLF and identify differentially expressed genes related to PANoptosis. Enrichment analysis showed that PANoptosis-related pathways were up-regulated in ACLF. We screened out BAX from the intersection of pyroptosis, apoptosis, necroptosis, and DEGs. Secondly, we screened articles from literature databases related to PANoptosis and liver failure, and specific forms of PANoptosis were reported in different experimental models in vitro and in vivo. Secondly, we established a model of ACLF using carbon tetrachloride-induced liver fibrosis, followed by D-galactosamine and lipopolysaccharide joint acute attacks. A substantial release of inflammatory factors(IL-6, IL-18, TNFα, and IFNγ) and the key proteins of PANoptosis (NLRP3, CASP1, GSDMD, BAX, CASP8, CASP3, CASP7, and MLKL) were detected independently in the ACLF rats. Finally, we found that combining TNF-α/INF-γ inflammatory cytokines could induce L02 cells PANoptosis. Our study highlighted the potential role of ACLF and helps drug discovery targeting PANoptosis in the future.
Collapse
Affiliation(s)
- Qianling Ye
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of TCM Collateral Disease Theory Research, Beijing, 100069, China
| | - Hanjing Wang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of TCM Collateral Disease Theory Research, Beijing, 100069, China
| | - Yue Chen
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of TCM Collateral Disease Theory Research, Beijing, 100069, China
| | - Yihao Zheng
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of TCM Collateral Disease Theory Research, Beijing, 100069, China
| | - Yuqiong Du
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China.
- Beijing Key Laboratory of TCM Collateral Disease Theory Research, Beijing, 100069, China.
| | - Chongyang Ma
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China.
- Beijing Key Laboratory of TCM Collateral Disease Theory Research, Beijing, 100069, China.
| | - Qiuyun Zhang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China.
- Beijing Key Laboratory of TCM Collateral Disease Theory Research, Beijing, 100069, China.
| |
Collapse
|
49
|
Puengel T, Tacke F. Role of Kupffer cells and other immune cells. SINUSOIDAL CELLS IN LIVER DISEASES 2024:483-511. [DOI: 10.1016/b978-0-323-95262-0.00024-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
50
|
Szafranska K, Sørensen KK, Lalor PF, McCourt P. Sinusoidal cells and liver immunology. SINUSOIDAL CELLS IN LIVER DISEASES 2024:53-75. [DOI: 10.1016/b978-0-323-95262-0.00003-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|