Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.033
1.
Nature ; 626(8000): 864-873, 2024 Feb.
Article En | MEDLINE | ID: mdl-38326607

Macrophage activation is controlled by a balance between activating and inhibitory receptors1-7, which protect normal tissues from excessive damage during infection8,9 but promote tumour growth and metastasis in cancer7,10. Here we report that the Kupffer cell lineage-determining factor ID3 controls this balance and selectively endows Kupffer cells with the ability to phagocytose live tumour cells and orchestrate the recruitment, proliferation and activation of natural killer and CD8 T lymphoid effector cells in the liver to restrict the growth of a variety of tumours. ID3 shifts the macrophage inhibitory/activating receptor balance to promote the phagocytic and lymphoid response, at least in part by buffering the binding of the transcription factors ELK1 and E2A at the SIRPA locus. Furthermore, loss- and gain-of-function experiments demonstrate that ID3 is sufficient to confer this potent anti-tumour activity to mouse bone-marrow-derived macrophages and human induced pluripotent stem-cell-derived macrophages. Expression of ID3 is therefore necessary and sufficient to endow macrophages with the ability to form an efficient anti-tumour niche, which could be harnessed for cell therapy in cancer.


Inhibitor of Differentiation Proteins , Kupffer Cells , Neoplasms , Animals , Humans , Mice , Bone Marrow Cells/cytology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Lineage , Induced Pluripotent Stem Cells/cytology , Inhibitor of Differentiation Proteins/deficiency , Inhibitor of Differentiation Proteins/genetics , Inhibitor of Differentiation Proteins/metabolism , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Kupffer Cells/cytology , Kupffer Cells/immunology , Kupffer Cells/metabolism , Liver/immunology , Liver/pathology , Macrophage Activation , Neoplasm Proteins , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Phagocytosis
3.
Science ; 381(6662): eabq5202, 2023 09 08.
Article En | MEDLINE | ID: mdl-37676943

Kupffer cells (KCs) are localized in liver sinusoids but extend pseudopods to parenchymal cells to maintain their identity and serve as the body's central bacterial filter. Liver cirrhosis drastically alters vascular architecture, but how KCs adapt is unclear. We used a mouse model of liver fibrosis and human tissue to examine immune adaptation. Fibrosis forced KCs to lose contact with parenchymal cells, down-regulating "KC identity," which rendered them incapable of clearing bacteria. Commensals stimulated the recruitment of monocytes through CD44 to a spatially distinct vascular compartment. There, recruited monocytes formed large aggregates of multinucleated cells (syncytia) that expressed phenotypical KC markers and displayed enhanced bacterial capture ability. Syncytia formed via CD36 and were observed in human cirrhosis as a possible antimicrobial defense that evolved with fibrosis.


Blood-Borne Infections , Giant Cells , Kupffer Cells , Liver Cirrhosis , Animals , Humans , Mice , Giant Cells/immunology , Giant Cells/microbiology , Kupffer Cells/immunology , Kupffer Cells/microbiology , Liver Cirrhosis/immunology , Liver Cirrhosis/microbiology , Liver Cirrhosis/pathology , Blood-Borne Infections/immunology , Disease Models, Animal
4.
Int J Cancer ; 152(12): 2615-2628, 2023 06 15.
Article En | MEDLINE | ID: mdl-36912275

Due to a combination of rapid disease progression and the lack of curative treatment options, hepatocellular carcinoma (HCC) is one of the deadliest cancers worldwide. Infiltrated, monocyte-derived, tumor-associated macrophages are known to play a role in HCC pathogenesis, but the involvement of Kupffer cells (KCs) remains elusive. Here, we used the Clec4F-diphteria toxin receptor transgenic mouse model to specifically investigate the effect of KC depletion on HCC initiation, progression and neoplastic growth following liver resection. For this purpose, several HCC mouse models with varying underlying etiologies were used and partial hepatectomy was performed. Our results show that in HCC, developed on a fibrotic or non-alcoholic steatohepatitis background, depletion of embryonic KCs at the onset of HCC induction and the subsequent replacement by monocyte-derived KCs does not affect the tumor burden, tumor microenvironment or the phenotype of isolated KCs at end-stage disease. In non-chronic liver disease-associated diethylnitrosamine-induced HCC, ablation of Clec4F+ KCs did not alter tumor progression or neoplastic growth following liver resection. Our results show that temporal ablation of resident KCs does not impact HCC pathogenesis, neither in the induction phase nor in advanced disease, and indicate that bone marrow-derived KCs are able to swiftly repopulate the available KC niche and adopt their phenotype.


Carcinogenesis , Carcinoma, Hepatocellular , Kupffer Cells , Liver Neoplasms, Experimental , Liver Neoplasms , Tumor-Associated Macrophages , Kupffer Cells/immunology , Disease Progression , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/pathology , Animals , Mice , Liver Neoplasms, Experimental/immunology , Liver Neoplasms, Experimental/pathology , Monocyte-Macrophage Precursor Cells/immunology , Carcinogenesis/immunology , Carcinogenesis/pathology , Mice, Inbred C57BL , Male
5.
J Immunol ; 208(11): 2558-2572, 2022 06 01.
Article En | MEDLINE | ID: mdl-35562117

Kupffer cells (KCs), the largest tissue-resident macrophage population in the body, play a central role in maintaining a delicate balance between immune tolerance and immunity in the liver. However, the underlying molecular mechanism remains elusive. In this study, we show that KCs express high levels of miR-146a, which is under control of the PU.1 transcription factor. miR-146a deficiency promoted KCs differentiation toward a proinflammatory phenotype; conversely, miR-146a overexpression suppressed this phenotypic differentiation. We found that hepatitis B virus (HBV) persistence or HBV surface Ag treatment significantly upregulated miR-146a expression and thereby impaired polarization of KCs toward a proinflammatory phenotype. Furthermore, in an HBV carrier mouse model, KCs depletion by clodronate liposomes dramatically promoted HBV clearance and enhanced an HBV-specific hepatic CD8+ T cell and CD4+ T cell response. Consistent with this finding, miR-146a knockout mice cleared HBV faster and elicited a stronger adaptive antiviral immunity than wild-type mice. In vivo IL-12 blockade promoted HBV persistence and tempered the HBV-specific CTL response in the liver of miR-146a knockout mice. Taken together, our results identified miR-146a as a critical intrinsic regulator of an immunosuppressive phenotype in KCs under inflammatory stimuli, which may be beneficial in maintenance of liver homeostasis under physiological condition. Meanwhile, during HBV infection, miR-146a contributed to viral persistence by inhibiting KCs proinflammatory polarization, highlighting its potential as a therapeutic target in HBV infection.


Hepatitis B , Immune Tolerance , Kupffer Cells , MicroRNAs , Animals , Hepatitis B/immunology , Hepatitis B virus , Kupffer Cells/immunology , Mice , Mice, Knockout , MicroRNAs/genetics
6.
Viruses ; 14(2)2022 01 24.
Article En | MEDLINE | ID: mdl-35215812

The role of non-parenchymal liver cells as part of the hepatic, innate immune system in the defense against hepatotropic viruses is not well understood. Here, primary human Kupffer cells, liver sinusoidal endothelial cells and hepatic stellate cells were isolated from liver tissue obtained after tumor resections or liver transplantations. Cells were stimulated with Toll-like receptor 1-9 ligands for 6-24 h. Non-parenchymal liver cells expressed and secreted inflammatory cytokines (IL6, TNF and IL10). Toll-like receptor- and cell type-specific downstream signals included the phosphorylation of NF-κB, AKT, JNK, p38 and ERK1/2. However, only supernatants of TLR3-activated Kupffer cells, liver sinusoidal endothelial cells and hepatic stellate cells contained type I and type III interferons and mediated an antiviral activity in the interferon-sensitive subgenomic hepatitis C virus replicon system. The antiviral effect could not be neutralized by antibodies against IFNA, IFNB nor IFNL, but could be abrogated using an interferon alpha receptor 2-specific neutralization. Interestingly, TLR3 responsiveness was enhanced in liver sinusoidal endothelial cells isolated from hepatitis C virus-positive donors, compared to uninfected controls. In conclusion, non-parenchymal liver cells are potent activators of the hepatic immune system by mediating inflammatory responses. Furthermore, liver sinusoidal endothelial cells were identified to be hyperresponsive to viral stimuli in chronic hepatitis C virus infection.


Hepacivirus/physiology , Hepatitis C, Chronic/immunology , Toll-Like Receptor 3/immunology , Animals , Endothelial Cells/immunology , Endothelial Cells/virology , Hepacivirus/genetics , Hepacivirus/immunology , Hepatic Stellate Cells/immunology , Hepatic Stellate Cells/virology , Hepatitis C, Chronic/genetics , Hepatitis C, Chronic/virology , Humans , Interferons/genetics , Interferons/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Kupffer Cells/immunology , Kupffer Cells/virology , Liver/immunology , Liver/virology , Male , Mice , Mice, Inbred C57BL , Toll-Like Receptor 3/genetics , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology
7.
Gastroenterology ; 162(2): 575-589, 2022 02.
Article En | MEDLINE | ID: mdl-34678217

BACKGROUND & AIMS: Hepatocellular carcinoma (HCC) is characterized by intratumoral accumulation of regulatory T cells (Tregs), which suppresses antitumor immunity. This study was designed to investigate how microRNAs regulate immunosuppression in HCC. METHODS: FVB/NJ mice were hydrodynamically injected with AKT/Ras or c-Myc and Sleeping Beauty transposon to induce HCC. The Sleeping Beauty system was used to deliver microRNA-15a/16-1 into livers of mice. Flow cytometry and immunostaining were used to determine changes in the immune system. RESULTS: Hydrodynamic injection of AKT/Ras or c-Myc into mice resulted in hepatic enrichment of Tregs and reduced cytotoxic T cells (CTLs) and HCC development. HCC impaired microRNA-15a/16-1 biogenesis in Kupffer cells (KCs) of AKT/Ras and c-Myc mice. Hydrodynamic injection of microRNA-15a/16-1 fully prevented HCC in AKT/Ras and c-Myc mice, while 100% of control mice died of HCC. Therapeutically, microRNA-15a/16-1 promoted a regression of HCC in both mouse models, impaired hepatic enrichment of Tregs, and increased hepatic CTLs. Mechanistically, a significant increase was observed in serum C-C motif chemokine 22 (CCL22) and transcription of Ccl22 in KCs of AKT/Ras and c-Myc mice. MicroRNA-15a/16-1 prevented KCs from overproducing CCL22 by inhibiting nuclear factor-κB that activates transcription of Ccl22. By reducing CCL22 binding to C-C chemokine receptor type 4 on Tregs, microRNA-15a/16-1 impaired Treg chemotaxis. Disrupting the interaction between microRNA-15a/16-1 and nuclear factor-κB impaired the ability of microRNA-15a/16-1 to prevent hepatic Treg accumulation and HCC. Depletion of cluster of differentiation 8+ T cells and additional treatment of CCL22 recovered growth of HCC that was fully prevented by microRNA-15a/16. CONCLUSIONS: MicroRNA-15a/16-1 attenuates immunosuppression by disrupting CCL22-mediated communication between KCs and Tregs. MicroRNA-15a/16-1 represents a potential immunotherapy against HCC.


Carcinoma, Hepatocellular/immunology , Kupffer Cells/immunology , Liver Neoplasms, Experimental/immunology , MicroRNAs/genetics , T-Lymphocytes, Regulatory/immunology , Tumor Escape/immunology , Animals , Carcinoma, Hepatocellular/genetics , Kupffer Cells/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Liver Neoplasms, Experimental/genetics , Mice , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-myc , T-Lymphocytes, Regulatory/metabolism , Tumor Escape/genetics , ras Proteins
8.
Int J Mol Sci ; 22(22)2021 Nov 10.
Article En | MEDLINE | ID: mdl-34830050

Many of the effector functions of antibodies rely on the binding of antibodies/immune complexes to cellular Fcγ receptors (FcγRs). Since the majority of innate immune effector cells express both activating and inhibitory Fc receptors, the outcome of the binding of immune complexes to cells of a given population is influenced by the relative affinities of the respective IgG subclasses to these receptors, as well as by the numbers of activating and inhibitory FcγRs on the cell surface. A group of immune cells that has come into focus more recently is the various subsets of tissue-resident macrophages. The central functions of FcγRs on tissue macrophages include the clearance of opsonized pathogens, the removal of small immune complexes from the circulation and the depletion of antibody-opsonized cells in the therapy of autoimmunity and cancer. Despite these essential functions of FcγRs on tissue-resident macrophages, an in-depth quantification of FcγRs is lacking. Thus, the aim of our current study was to quantify the various Fcγ receptors on macrophages in murine liver, lung, kidney, brain, skin and spleen. Our study identified a pronounced heterogeneity between FcγR expression patterns of the different tissue macrophages, which may reflect their specialized functions within their unique niches in different organ environments.


Macrophages/immunology , Macrophages/metabolism , Receptors, IgG/immunology , Receptors, IgG/metabolism , Animals , Biomarkers/metabolism , Female , Kidney/immunology , Kidney/metabolism , Kupffer Cells/immunology , Kupffer Cells/metabolism , Lung/immunology , Lung/metabolism , Mice , Microglia/immunology , Microglia/metabolism , Receptors, IgG/analysis , Skin/immunology , Skin/metabolism , Spleen/immunology , Spleen/metabolism
9.
Int J Biol Macromol ; 193(Pt B): 1871-1884, 2021 Dec 15.
Article En | MEDLINE | ID: mdl-34774589

This study is to investigate the effect of microRNA-1338 (miR-1338) on the activity of Kupffer cells (KCs) and its mechanism regulated by ophiopogon polysaccharide liposome (OPL). KCs was treated with different OPL after transfected with miR-1338 mimic and miR-1338 inhibitor. The secretion of NO and iNOS, the expression of catalase (CAT) and peroxidase (POD), the phagocytic activity, the expression of CD14 and MHC II, the apoptosis and the secretion of ROS were measured. In addition, the expressions of key signal factors TLR4, IKKß, MyD88 and NF-κB in NF-κB signaling pathway were measured by real-time PCR and Western blot (WB). The results showed that OPL could promote the secretion of iNOS, the expression of POD, the phagocytosis, the mRNA expression of TLR4, MyD88, IKKß and NF-κB, the protein expression of TLR4 and NF-κB, and inhibit the cell apoptosis and ROS secretion after transfected with miR-1338 mimic. After transfected with miR-1338 inhibitor, OPL could promote the secretion of NO and iNOS, the expression of POD, cell migration, phagocytosis, and inhibit cell apoptosis. Meanwhile, the mRNA expression of TLR4, MyD88, IKKß and NF-κB and the protein expression of TLR4, MyD88 and NF-κB were promoted. These results suggested that OPL could activate TLR4-NF-κB signaling pathway and thereby improve the activity of KCs by regulating miR-1338.


Immunologic Factors , Kupffer Cells/immunology , MicroRNAs/immunology , Ophiopogon/chemistry , Polysaccharides , Animals , Immunologic Factors/chemistry , Immunologic Factors/pharmacology , Liposomes , Mice , Polysaccharides/chemistry , Polysaccharides/pharmacology
10.
Front Immunol ; 12: 654998, 2021.
Article En | MEDLINE | ID: mdl-34531848

HCV core protein is the first structural protein synthesized during hepatitis C virus (HCV) infection and replication. It is released from virus infected liver cells and mediates multiple functions to affect host cell response. The innate immune response is the first line of defense against viral infection. After HCV infection, Kupffer cells (KCs) which are liver macrophages play an important role in host innate immune response. Kupffer cells act as phagocytes and release different cytokines and chemokines to counter viral infection and regulate inflammation and fibrosis in liver. Earlier, we have demonstrated that HCV core protein interacts with gC1qR and activates MAPK, NF-κB and PI3K/AKT pathways in macrophages. In this study, we explored the effect of HCV core protein on CCL2 and CXCL10 expression in macrophages and the signaling pathways involved. Upon silencing of gC1qR, we observed a significant decrease expression of CCL2 and CXCL10 in macrophages in the presence of HCV core protein. Inhibiting NF-κB pathway, but not P38, JNK, ERK and AKT pathways greatly reduced the expression of CCL2 and CXCL10. Therefore, our results indicate that interaction of HCV core protein with gC1qR could induce CCL2 and CXCL10 secretion in macrophages via NF-κB signaling pathway. These findings may shed light on the understanding of how leukocytes migrate into the liver and exaggerate host-derived immune responses and may provide novel therapeutic targets in HCV chronic inflammation.


Chemokine CCL2/immunology , Chemokine CXCL10/immunology , Hepacivirus/immunology , Macrophages/immunology , NF-kappa B/immunology , Signal Transduction/immunology , Viral Core Proteins/immunology , Animals , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Gene Expression/immunology , Hepacivirus/metabolism , Hepacivirus/physiology , Hepatitis C/immunology , Hepatitis C/metabolism , Hepatitis C/virology , Host-Pathogen Interactions/immunology , Humans , Kupffer Cells/immunology , Kupffer Cells/metabolism , Kupffer Cells/virology , Macrophages/metabolism , Macrophages/virology , Mice , Mice, Inbred BALB C , RAW 264.7 Cells , THP-1 Cells , Viral Core Proteins/metabolism
11.
Cell Prolif ; 54(10): e13116, 2021 Oct.
Article En | MEDLINE | ID: mdl-34469018

OBJECTIVES: The discrepancy between supply and demand of organ has led to an increased utilization of steatotic liver for liver transplantation (LT). Hepatic steatosis, however, is a major risk factor for graft failure due to increased susceptibility to ischaemia-reperfusion (I/R) injury during transplantation. MATERIALS AND METHODS: To assess the plasticity and phenotype of immune cells within the microenvironment of steatotic liver graft at single-cell level, single-cell RNA-sequencing (scRNA-Seq) was carried out on 23 675 cells from transplanted rat livers. Bioinformatic analyses and multiplex immunohistochemistry were performed to assess the functional properties, transcriptional regulation, phenotypic switching and cell-cell interactions of different cell subtypes. RESULTS: We have identified 11 different cell types in transplanted livers and found that the highly complex ecosystem was shaped by myeloid-derived cell subsets that transit between different states and interact mutually. Notably, a pro-inflammatory phenotype of Kupffer cells (KCs) with high expression of colony-stimulating factor 3 (CSF3) that was enriched in transplanted steatotic livers was potentially participated in fatty graft injury. We have also detected a subset of dendritic cells (DCs) with highly expressing XCR1 that was correlated with CD8+ T cells, mediating the severer steatotic liver damage by I/R injury. CONCLUSIONS: The findings of our study provide new insight into the mechanisms by which steatosis exacerbates liver damage from I/R injury. Interventions based on these observations create opportunities in attenuating fatty liver graft injury and expanding the donor pool.


Fatty Liver/immunology , Liver/immunology , Reperfusion Injury/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Communication/immunology , Disease Models, Animal , Kupffer Cells/immunology , Liver Transplantation/methods , Phenotype , Rats , Rats, Sprague-Dawley , Single-Cell Analysis/methods , Transcription, Genetic/immunology
12.
Immunity ; 54(9): 2089-2100.e8, 2021 09 14.
Article En | MEDLINE | ID: mdl-34469774

Kupffer cells (KCs) are highly abundant, intravascular, liver-resident macrophages known for their scavenger and phagocytic functions. KCs can also present antigens to CD8+ T cells and promote either tolerance or effector differentiation, but the mechanisms underlying these discrepant outcomes are poorly understood. Here, we used a mouse model of hepatitis B virus (HBV) infection, in which HBV-specific naive CD8+ T cells recognizing hepatocellular antigens are driven into a state of immune dysfunction, to identify a subset of KCs (referred to as KC2) that cross-presents hepatocellular antigens upon interleukin-2 (IL-2) administration, thus improving the antiviral function of T cells. Removing MHC-I from all KCs, including KC2, or selectively depleting KC2 impaired the capacity of IL-2 to revert the T cell dysfunction induced by intrahepatic priming. In summary, by sensing IL-2 and cross-presenting hepatocellular antigens, KC2 overcome the tolerogenic potential of the hepatic microenvironment, suggesting new strategies for boosting hepatic T cell immunity.


Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , Cross-Priming/immunology , Interleukin-2/immunology , Kupffer Cells/immunology , Animals , Hepatitis B/immunology , Immune Tolerance/immunology , Mice , Mice, Transgenic
13.
Int J Mol Sci ; 22(15)2021 Jul 27.
Article En | MEDLINE | ID: mdl-34360773

Non-alcoholic fatty liver disease (NAFLD) is a leading cause of liver cirrhosis and hepatocellular carcinoma. NAFLD is associated with metabolic disorders such as obesity, insulin resistance, dyslipidemia, steatohepatitis, and liver fibrosis. Liver-resident (Kupffer cells) and recruited macrophages contribute to low-grade chronic inflammation in various tissues by modulating macrophage polarization, which is implicated in the pathogenesis of metabolic diseases. Abnormalities in the intestinal environment, such as the gut microbiota, metabolites, and immune system, are also involved in the pathogenesis and development of NAFLD. Hepatic macrophage activation is induced by the permeation of antigens, endotoxins, and other proinflammatory substances into the bloodstream as a result of increased intestinal permeability. Therefore, it is important to understand the role of the gut-liver axis in influencing macrophage activity, which is central to the pathogenesis of NAFLD and nonalcoholic steatohepatitis (NASH). Not only probiotics but also biogenics (heat-killed lactic acid bacteria) are effective in ameliorating the progression of NASH. Here we review the effect of hepatic macrophages/Kupffer cells, other immune cells, intestinal permeability, and immunity on NAFLD and NASH and the impact of probiotics, prebiotics, and biogenesis on those diseases.


Gastrointestinal Microbiome/immunology , Non-alcoholic Fatty Liver Disease , Prebiotics , Probiotics , Animals , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/microbiology , Humans , Kupffer Cells/immunology , Liver Neoplasms/immunology , Liver Neoplasms/microbiology , Non-alcoholic Fatty Liver Disease/immunology , Non-alcoholic Fatty Liver Disease/microbiology
14.
Science ; 373(6553)2021 07 23.
Article En | MEDLINE | ID: mdl-34437091

The biogenesis of high-density lipoprotein (HDL) requires apoA1 and the cholesterol transporter ABCA1. Although the liver generates most of the HDL in the blood, HDL synthesis also occurs in the small intestine. Here, we show that intestine-derived HDL traverses the portal vein in the HDL3 subspecies form, in complex with lipopolysaccharide (LPS)-binding protein (LBP). HDL3, but not HDL2 or low-density lipoprotein, prevented LPS binding to and inflammatory activation of liver macrophages and instead supported extracellular inactivation of LPS. In mouse models involving surgical, dietary, or alcoholic intestinal insult, loss of intestine-derived HDL worsened liver injury, whereas outcomes were improved by therapeutics that elevated and depended upon raising intestinal HDL. Thus, protection of the liver from injury in response to gut-derived LPS is a major function of intestinally synthesized HDL.


Intestine, Small/metabolism , Lipoproteins, HDL3/metabolism , Liver Diseases/prevention & control , Liver/metabolism , Portal Vein/metabolism , Acute-Phase Proteins/metabolism , Adult , Animals , Carrier Proteins/metabolism , Cholesterol, HDL/blood , Cholesterol, HDL/metabolism , Enterocytes/metabolism , Humans , Intestine, Small/surgery , Kupffer Cells/immunology , Kupffer Cells/metabolism , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/metabolism , Lipoproteins, HDL3/blood , Liver/pathology , Liver Cirrhosis/pathology , Liver Cirrhosis/prevention & control , Liver Diseases/pathology , Liver X Receptors/metabolism , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Protein Binding , Signal Transduction , Toll-Like Receptor 4/metabolism
15.
Cells ; 10(8)2021 07 27.
Article En | MEDLINE | ID: mdl-34440668

Considering that the heterogenic population of a hepatic progenitor cell line (HPCL) can play a vital role in autoimmune hepatitis (AIH), we decided to conduct pioneering retrospective evaluation of these cells in pediatric AIH by means of transmission electron microscopy (TEM). The aim of the study was to assess the ultrastructure of the HPCL in children with untreated AIH. Ultrastructural analysis of the HPCL population, preceded by immunohistochemical staining for cytokeratin 7 (CK7), was performed using pretreatment liver biopsies from 23 children with clinicopathologically diagnosed AIH. Immunohistochemical assessment for CK7 allowed detection of proliferating immature epithelial cells differentiating towards periportal and intralobular intermediate hepatocytes without marked formation of ductular reactions in AIH children. Using TEM, we distinguished three morphological types of HPCs: I-the most undifferentiated progenitor cells; III-intermediate hepatocyte-like cells; II-intermediate bile duct cells. Most frequent were the cells differentiating towards hepatocytes, most rare-those differentiating towards cholangiocytes. The results indicate that an HPCL may be an important source of hepatocyte regeneration. Ultrastructural analyses of the HPCL population, combined with immunohistochemistry for CK7, might be a useful tool to evaluate liver cell regeneration, including fibrogenesis, and may help better understand the morphological pattern of the disease, in pediatric AIH. Frequent appearance of an HPCL in the vicinity of fibrotic foci, often accompanied by hyperactive Kupffer cells and transitional hepatic stellate cells, may indicate their significant involvement in liver fibrogenesis.


Hepatitis, Autoimmune/metabolism , Immunohistochemistry , Keratin-7/metabolism , Liver Regeneration , Liver/metabolism , Microscopy, Electron, Transmission , Stem Cells/metabolism , Adolescent , Age Factors , Biomarkers/blood , Cell Differentiation , Cell Line , Cell Proliferation , Child , Child, Preschool , Female , Hepatitis, Autoimmune/immunology , Hepatitis, Autoimmune/pathology , Hepatocytes/immunology , Hepatocytes/metabolism , Hepatocytes/ultrastructure , Humans , Kupffer Cells/immunology , Kupffer Cells/metabolism , Kupffer Cells/ultrastructure , Liver/immunology , Liver/ultrastructure , Male , Predictive Value of Tests , Retrospective Studies , Stem Cells/immunology , Stem Cells/ultrastructure
16.
Front Immunol ; 12: 697362, 2021.
Article En | MEDLINE | ID: mdl-34234785

Hepatic injury induced by ischemia and reperfusion (HIRI) is a major clinical problem after liver resection or transplantation. The polarization of macrophages plays an important role in regulating the severity of hepatic ischemia/reperfusion injury. Recent evidence had indicated that the ischemia induces an acidic microenvironment by causing increased anaerobic glycolysis and accumulation of lactic acid. We hypothesize that the acidic microenvironment might cause the imbalance of intrahepatic immunity which aggravated HIRI. The hepatic ischemia/reperfusion injury model was established to investigate the effect of the acidic microenvironment to liver injury. Liposomes were used to deplete macrophages in vivo. Macrophages were cultured under low pH conditions to analyze the polarization of macrophages in vitro. Activation of the PPAR-γ signal was determined by Western blot. PPAR-γ agonist GW1929 was administrated to functionally test the role of PPAR-γ in regulating macrophage-mediated effects in the acidic microenvironment during HIRI. We demonstrate that acidic microenvironment aggravated HIRI while NaHCO3 reduced liver injury through neutralizing the acid, besides, liposome abolished the protective ability of NaHCO3 through depleting the macrophages. In vivo and vitro experiment showed that acidic microenvironment markedly promoted M1 polarization but inhibited M2 polarization of macrophage. Furthermore, the mechanistic study proved that the PPAR-γ signal was suppressed during the polarization of macrophages under pH = 6.5 culture media. The addition of PPAR-γ agonist GW1929 inhibited M1 polarization under acidic environment and reduced HIRI. Our results indicate that acidic microenvironment is a key regulator in HIRI which promoted M1 polarization of macrophages through regulating PPAR-γ. Conversely, PPAR-γ activation reduced liver injury, which provides a novel therapeutic concept to prevent HIRI.


Liver/injuries , Liver/metabolism , Macrophages/metabolism , PPAR gamma/metabolism , Reperfusion Injury/metabolism , Animals , Benzophenones/administration & dosage , Cells, Cultured , Cellular Microenvironment/drug effects , Cellular Microenvironment/physiology , Disease Models, Animal , Hydrogen-Ion Concentration , Immunity, Innate/drug effects , Kupffer Cells/drug effects , Kupffer Cells/immunology , Kupffer Cells/metabolism , Liver/pathology , Macrophages/classification , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , PPAR gamma/agonists , Reperfusion Injury/drug therapy , Reperfusion Injury/pathology , Severity of Illness Index , Signal Transduction , Sodium Bicarbonate/pharmacology , Tyrosine/administration & dosage , Tyrosine/analogs & derivatives
17.
Sci Immunol ; 6(61)2021 07 02.
Article En | MEDLINE | ID: mdl-34215680

Immunotherapy is revolutionizing cancer treatment but is often restricted by toxicities. What distinguishes adverse events from concomitant antitumor reactions is poorly understood. Here, using anti-CD40 treatment in mice as a model of TH1-promoting immunotherapy, we showed that liver macrophages promoted local immune-related adverse events. Mechanistically, tissue-resident Kupffer cells mediated liver toxicity by sensing lymphocyte-derived IFN-γ and subsequently producing IL-12. Conversely, dendritic cells were dispensable for toxicity but drove tumor control. IL-12 and IFN-γ were not toxic themselves but prompted a neutrophil response that determined the severity of tissue damage. We observed activation of similar inflammatory pathways after anti-PD-1 and anti-CTLA-4 immunotherapies in mice and humans. These findings implicated macrophages and neutrophils as mediators and effectors of aberrant inflammation in TH1-promoting immunotherapy, suggesting distinct mechanisms of toxicity and antitumor immunity.


Immune Checkpoint Inhibitors/adverse effects , Immunotherapy/adverse effects , Kupffer Cells/drug effects , Liver/drug effects , Neoplasms/therapy , Neutrophils/drug effects , Animals , CD40 Antigens/antagonists & inhibitors , CD40 Antigens/immunology , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/immunology , Cytokines/immunology , Humans , Kupffer Cells/immunology , Liver/immunology , Mice, Transgenic , Neoplasms/immunology , Neutrophils/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology
18.
PLoS One ; 16(7): e0254886, 2021.
Article En | MEDLINE | ID: mdl-34297734

In the murine liver, there are two major macrophage populations, namely resident Kupffer cells (resKCs) with phagocytic activity and recruited macrophages (recMφs) with cytokine-producing capacity. This study was performed to clarify the functional differences between these two populations, focusing on their susceptibility to radiation and response to stimulation via liver X receptors (LXRs), which are implicated in cholesterol metabolism and immune regulation. Liver mononuclear cells (MNCs) were obtained from C57BL/6 (WT) mice with or without 2 Gy irradiation, and the phagocytic activity against Escherichia coli (E. coli) as well as TNF-α production were compared between the two macrophage populations. To assess LXR functions, phagocytosis, TNF-α production, and endocytosis of acetylated low-density lipoprotein (LDL) were compared after synthetic LXR ligand stimulation. Furthermore, LXRα/ß knockout (KO) mice and LXRα KO mice were compared with WT mice. Irradiation decreased intracellular TNF-α production by recMφs but did not affect the phagocytic activity of resKCs. In vitro LXR stimulation enhanced E. coli phagocytosis by resKCs but decreased E. coli-stimulated TNF-α production by recMφs. Phagocytic activity and acetylated LDL endocytosis were decreased in both LXRα/ß KO mice and LXRα KO mice, with serum TNF-α levels after E. coli injection in the former being higher than those in WT mice. In conclusion, resKCs and recMφs exhibited different functional features in response to radiation and LXR stimulation, highlighting their distinct roles liver immunity and lipid metabolism.


Kupffer Cells/immunology , Liver X Receptors/metabolism , Liver/immunology , Phagocytosis , Animals , Cells, Cultured , Lipid Metabolism , Lipoproteins, LDL/metabolism , Liver/cytology , Liver/radiation effects , Liver X Receptors/genetics , Mice , Mice, Inbred C57BL , Tumor Necrosis Factor-alpha/metabolism
19.
Nat Metab ; 3(6): 843-858, 2021 06.
Article En | MEDLINE | ID: mdl-34127858

Pre-operative exercise therapy improves outcomes for many patients who undergo surgery. Despite the well-known effects on tolerance to systemic perturbation, the mechanisms by which pre-operative exercise protects the organ that is operated on from inflammatory injury are unclear. Here, we show that four-week aerobic pre-operative exercise significantly attenuates liver injury and inflammation from ischaemia and reperfusion in mice. Remarkably, these beneficial effects last for seven more days after completing pre-operative exercising. We find that exercise specifically drives Kupffer cells toward an anti-inflammatory phenotype with trained immunity via metabolic reprogramming. Mechanistically, exercise-induced HMGB1 release enhances itaconate metabolism in the tricarboxylic acid cycle that impacts Kupffer cells in an NRF2-dependent manner. Therefore, these metabolites and cellular/molecular targets can be investigated as potential exercise-mimicking pharmaceutical candidates to protect against liver injury during surgery.


Energy Metabolism , Immunity, Innate , Kupffer Cells/immunology , Kupffer Cells/metabolism , Preoperative Exercise , Animals , Disease Resistance , Inflammation/immunology , Inflammation/metabolism , Ischemia/immunology , Ischemia/metabolism , Mice
20.
Front Immunol ; 12: 671073, 2021.
Article En | MEDLINE | ID: mdl-34012454

The liver is the central organ for cholesterol synthesis and homeostasis. The effects of dietary cholesterol on hepatic injury, mainly of oxidized low-density lipoproteins (OxLDL), are not fully understood. Here, we show that the degree of cholesterol oxidation had different impacts on the global gene expression of human M2-like macrophages, with highly oxidized LDL causing the most dramatic changes. M2-like macrophages and Kupffer cells undergo M4-like polarization, decreasing the expression of important markers, such as IL10, MRC1, and CD163. These cells also displayed functional changes, with reduced phagocytic capacity, increased neutrophil recruitment, and more effective neutrophil extracellular traps (NETs) induction. Our findings provide a link between LDL oxidation and modification of peripheral and liver macrophage function.


Cholesterol/metabolism , Extracellular Traps/metabolism , Kupffer Cells/immunology , Macrophages/immunology , Neutrophils/physiology , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Cell Differentiation , Cells, Cultured , Gene Expression Regulation , Humans , Interleukin-10/metabolism , Membrane Glycoproteins , Phagocytosis , Receptors, Cell Surface/metabolism , Receptors, Immunologic
...