Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 12.557
Filter
1.
J Nanobiotechnology ; 22(1): 315, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38840207

ABSTRACT

Chronic hepatitis B poses a significant global burden, modulating immune cells, leading to chronic inflammation and long-term damage. Due to its hepatotropism, the hepatitis B virus (HBV) cannot infect other cells. The mechanisms underlying the intercellular communication among different liver cells in HBV-infected individuals and the immune microenvironment imbalance remain elusive. Exosomes, as important intercellular communication and cargo transportation tools between HBV-infected hepatocytes and immune cells, have been shown to assist in HBV cargo transportation and regulate the immune microenvironment. However, the role of exosomes in hepatitis B has only gradually received attention in recent years. Minimal literature has systematically elaborated on the role of exosomes in reshaping the immune microenvironment of the liver. This review unfolds sequentially based on the biological processes of exosomes: exosomes' biogenesis, release, transport, uptake by recipient cells, and their impact on recipient cells. We delineate how HBV influences the biogenesis of exosomes, utilizing exosomal covert transmission, and reshapes the hepatic immune microenvironment. And based on the characteristics and functions of exosomes, potential applications of exosomes in hepatitis B are summarized and predicted.


Subject(s)
Exosomes , Hepatitis B virus , Hepatitis B, Chronic , Hepatocytes , Liver , Exosomes/immunology , Exosomes/metabolism , Humans , Hepatitis B virus/immunology , Liver/immunology , Liver/virology , Animals , Hepatitis B, Chronic/immunology , Hepatocytes/virology , Hepatocytes/immunology , Cell Communication , Cellular Microenvironment/immunology , Hepatitis B/immunology , Hepatitis B/virology
2.
Parasites Hosts Dis ; 62(2): 243-250, 2024 May.
Article in English | MEDLINE | ID: mdl-38835265

ABSTRACT

We investigated organ specific Toxocara canis larval migration in mice infected with T. canis larvae. We observed the worm burden and systemic immune responses. Three groups of BALB/c mice (n=5 each) were orally administered 1,000 T. canis 2nd stage larvae to induce larva migrans. Mice were sacrificed at 1, 3, and 5 weeks post-infection. Liver, lung, brain, and eye tissues were collected. Tissue from 2 mice per group was digested for larval count, while the remaining 3 mice underwent histological analysis. Blood hematology and serology were evaluated and compared to that in a control uninfected group (n=5) to assess the immune response. Cytokine levels in bronchoalveolar lavage (BAL) fluid were also analyzed. We found that, 1 week post-infection, the mean parasite load in the liver (72±7.1), brain (31±4.2), lungs (20±5.7), and eyes (2±0) peaked and stayed constant until the 3 weeks. By 5-week post-infection, the worm burden in the liver and lungs significantly decreased to 10±4.2 and 9±5.7, respectively, while they remained relatively stable in the brain and eyes (18±4.2 and 1±0, respectively). Interestingly, ocular larvae resided in all retinal layers, without notable inflammation in outer retina. Mice infected with T. canis exhibited elevated levels of neutrophils, monocytes, eosinophils, and immunoglobulin E. At 5 weeks post-infection, interleukin (IL)-5 and IL-13 levels were elevated in BAL fluid. Whereas IL-4, IL-10, IL-17, and interferon-γ levels in BAL fluid were similar to that in controls. Our findings demonstrate that a small portion of T. canis larvae migrate to the eyes and brain within the first week of infection. Minimal tissue inflammation was observed, probably due to increase of anti-inflammatory cytokines. This study contributes to our understanding of the histological and immunological responses to T. canis infection in mice, which may have implications to further understand human toxocariasis.


Subject(s)
Brain , Cytokines , Larva , Liver , Lung , Mice, Inbred BALB C , Toxocara canis , Toxocariasis , Animals , Toxocara canis/immunology , Toxocariasis/immunology , Toxocariasis/pathology , Toxocariasis/parasitology , Larva/immunology , Mice , Cytokines/metabolism , Lung/parasitology , Lung/immunology , Lung/pathology , Liver/parasitology , Liver/pathology , Liver/immunology , Brain/parasitology , Brain/immunology , Brain/pathology , Bronchoalveolar Lavage Fluid/immunology , Bronchoalveolar Lavage Fluid/parasitology , Female , Parasite Load , Eye/parasitology , Eye/immunology , Eye/pathology , Disease Models, Animal
3.
Front Immunol ; 15: 1279043, 2024.
Article in English | MEDLINE | ID: mdl-38840916

ABSTRACT

Schistosomiasis remains the most devastating neglected tropical disease, affecting over 240 million people world-wide. The disease is caused by the eggs laid by mature female worms that are trapped in host's tissues, resulting in chronic Th2 driven fibrogranulmatous pathology. Although the disease can be treated with a relatively inexpensive drug, praziquantel (PZQ), re-infections remain a major problem in endemic areas. There is a need for new therapeutic drugs and alternative drug treatments for schistosomiasis. The current study hypothesized that cysteinyl leukotrienes (cysLTs) could mediate fibroproliferative pathology during schistosomiasis. Cysteinyl leukotrienes (cysLTs) are potent lipid mediators that are known to be key players in inflammatory diseases, such as asthma and allergic rhinitis. The present study aimed to investigate the role of cysLTR1 during experimental acute and chronic schistosomiasis using cysLTR1-/- mice, as well as the use of cysLTR1 inhibitor (Montelukast) to assess immune responses during chronic Schistosoma mansoni infection. Mice deficient of cysLTR1 and littermate control mice were infected with either high or low dose of Schistosoma mansoni to achieve chronic or acute schistosomiasis, respectively. Hepatic granulomatous inflammation, hepatic fibrosis and IL-4 production in the liver was significantly reduced in mice lacking cysLTR1 during chronic schistosomiasis, while reduced liver pathology was observed during acute schistosomiasis. Pharmacological blockade of cysLTR1 using montelukast in combination with PZQ reduced hepatic inflammation and parasite egg burden in chronically infected mice. Combination therapy led to the expansion of Tregs in chronically infected mice. We show that the disruption of cysLTR1 is dispensable for host survival during schistosomiasis, suggesting an important role cysLTR1 may play during early immunity against schistosomiasis. Our findings revealed that the combination of montelukast and PZQ could be a potential prophylactic treatment for chronic schistosomiasis by reducing fibrogranulomatous pathology in mice. In conclusion, the present study demonstrated that cysLTR1 is a potential target for host-directed therapy to ameliorate fibrogranulomatous pathology in the liver during chronic and acute schistosomiasis in mice.


Subject(s)
Acetates , Cyclopropanes , Disease Models, Animal , Mice, Knockout , Quinolines , Receptors, Leukotriene , Schistosomiasis mansoni , Sulfides , Animals , Receptors, Leukotriene/metabolism , Mice , Cyclopropanes/therapeutic use , Cyclopropanes/pharmacology , Acetates/therapeutic use , Acetates/pharmacology , Sulfides/therapeutic use , Sulfides/pharmacology , Schistosomiasis mansoni/drug therapy , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/parasitology , Quinolines/therapeutic use , Quinolines/pharmacology , Female , Schistosoma mansoni/immunology , Chronic Disease , Leukotriene Antagonists/pharmacology , Leukotriene Antagonists/therapeutic use , Liver/parasitology , Liver/pathology , Liver/metabolism , Liver/immunology , Mice, Inbred C57BL , Praziquantel/therapeutic use , Praziquantel/pharmacology , T-Lymphocytes, Regulatory/immunology
4.
Front Immunol ; 15: 1381735, 2024.
Article in English | MEDLINE | ID: mdl-38840923

ABSTRACT

Background: Acute liver injury (ALI), which is a type of inflammation-mediated hepatocellular injury, is a clinical syndrome that results from hepatocellular apoptosis and hemorrhagic necrosis. Apoptosis stimulating protein of p53-2 (ASPP2) is a proapoptotic member of the p53 binding protein family. However, the role of ASPP2 in the pathogenesis of ALI and its regulatory mechanisms remain unclear. Methods: The expression of ASPP2 were compared between liver biopsies derived from patients with CHB, patients with ALI, and normal controls. Acute liver injury was modelled in mice by administration of D-GalN/LPS. Liver injury was demonstrated by serum transaminases and histological assessment of liver sections. ASPP2-knockdown mice (ASPP2+/-) were used to determine its role in acute liver injury. Mouse bone marrow macrophages (BMMs) were isolated from wildtype and ASPP2+/- mice and stimulated with LPS, and the supernatant was collected to incubate with the primary hepatocytes. Quantitative real-time PCR and western blot were used to analyze the expression level of target. Results: The expression of ASPP2 was significantly upregulated in the liver tissue of ALI patients and acute liver injury mice. ASPP2+/- mice significantly relieved liver injury through reducing liver inflammation and decreasing hepatocyte apoptosis. Moreover, the conditioned medium (CM) of ASPP2+/- bone marrow-derived macrophages (BMMs) protected hepatocytes against apoptosis. Mechanistically, we revealed that ASPP2 deficiency in BMMs specifically upregulated IL-6 through autophagy activation, which decreased the level of TNF-α to reduce hepatocytes apoptosis. Furthermore, up-regulation of ASPP2 sensitizes hepatocytes to TNF-α-induced apoptosis. Conclusion: Our novel findings show the critical role of ASPP2 in inflammatory immunoregulatory mechanism of ALI and provide a rationale to target ASPP2 as a refined therapeutic strategy to ameliorate acute liver injury.


Subject(s)
Apoptosis Regulatory Proteins , Apoptosis , Animals , Humans , Mice , Male , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Mice, Knockout , Liver/pathology , Liver/metabolism , Liver/immunology , Hepatocytes/metabolism , Hepatocytes/pathology , Mice, Inbred C57BL , Disease Models, Animal , Inflammation/immunology , Inflammation/metabolism , Female , Lipopolysaccharides , Middle Aged , Macrophages/immunology , Macrophages/metabolism , Adult , Tumor Suppressor Proteins
5.
Nat Commun ; 15(1): 4711, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38830841

ABSTRACT

The fetal development of organs and functions is vulnerable to perturbation by maternal inflammation which may increase susceptibility to disorders after birth. Because it is not well understood how the placenta and fetus respond to acute lung- inflammation, we characterize the response to maternal pulmonary lipopolysaccharide exposure across 24 h in maternal and fetal organs using multi-omics, imaging and integrative analyses. Unlike maternal organs, which mount strong inflammatory immune responses, the placenta upregulates immuno-modulatory genes, in particular the IL-6 signaling suppressor Socs3. Similarly, we observe no immune response in the fetal liver, which instead displays metabolic changes, including increases in lipids containing docosahexaenoic acid, crucial for fetal brain development. The maternal liver and plasma display similar metabolic alterations, potentially increasing bioavailability of docosahexaenoic acid for the mother and fetus. Thus, our integrated temporal analysis shows that systemic inflammation in the mother leads to a metabolic perturbation in the fetus.


Subject(s)
Fetus , Lipopolysaccharides , Liver , Lung , Placenta , Female , Pregnancy , Placenta/metabolism , Placenta/immunology , Animals , Fetus/immunology , Fetus/metabolism , Lung/immunology , Lung/metabolism , Liver/metabolism , Liver/immunology , Docosahexaenoic Acids/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism , Suppressor of Cytokine Signaling 3 Protein/genetics , Mice , Inflammation/immunology , Inflammation/metabolism , Mice, Inbred C57BL , Adaptation, Physiological/immunology , Fetal Development/immunology , Maternal-Fetal Exchange/immunology , Interleukin-6/metabolism , Interleukin-6/immunology
6.
JCO Clin Cancer Inform ; 8: e2300159, 2024 May.
Article in English | MEDLINE | ID: mdl-38728613

ABSTRACT

PURPOSE: We present and validate a rule-based algorithm for the detection of moderate to severe liver-related immune-related adverse events (irAEs) in a real-world patient cohort. The algorithm can be applied to studies of irAEs in large data sets. METHODS: We developed a set of criteria to define hepatic irAEs. The criteria include: the temporality of elevated laboratory measurements in the first 2-14 weeks of immune checkpoint inhibitor (ICI) treatment, steroid intervention within 2 weeks of the onset of elevated laboratory measurements, and intervention with a duration of at least 2 weeks. These criteria are based on the kinetics of patients who experienced moderate to severe hepatotoxicity (Common Terminology Criteria for Adverse Events grades 2-4). We applied these criteria to a retrospective cohort of 682 patients diagnosed with hepatocellular carcinoma and treated with ICI. All patients were required to have baseline laboratory measurements before and after the initiation of ICI. RESULTS: A set of 63 equally sampled patients were reviewed by two blinded, clinical adjudicators. Disagreements were reviewed and consensus was taken to be the ground truth. Of these, 25 patients with irAEs were identified, 16 were determined to be hepatic irAEs, 36 patients were nonadverse events, and two patients were of indeterminant status. Reviewers agreed in 44 of 63 patients, including 19 patients with irAEs (0.70 concordance, Fleiss' kappa: 0.43). By comparison, the algorithm achieved a sensitivity and specificity of identifying hepatic irAEs of 0.63 and 0.81, respectively, with a test efficiency (percent correctly classified) of 0.78 and outcome-weighted F1 score of 0.74. CONCLUSION: The algorithm achieves greater concordance with the ground truth than either individual clinical adjudicator for the detection of irAEs.


Subject(s)
Algorithms , Immune Checkpoint Inhibitors , Liver Neoplasms , Humans , Immune Checkpoint Inhibitors/adverse effects , Male , Female , Middle Aged , Aged , Liver Neoplasms/drug therapy , Liver Neoplasms/immunology , Retrospective Studies , Phenotype , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/diagnosis , Carcinoma, Hepatocellular/drug therapy , Drug-Related Side Effects and Adverse Reactions/diagnosis , Drug-Related Side Effects and Adverse Reactions/etiology , Liver/pathology , Liver/drug effects , Liver/immunology
7.
Front Immunol ; 15: 1372441, 2024.
Article in English | MEDLINE | ID: mdl-38690269

ABSTRACT

Background and aims: Cuproptosis has emerged as a significant contributor in the progression of various diseases. This study aimed to assess the potential impact of cuproptosis-related genes (CRGs) on the development of hepatic ischemia and reperfusion injury (HIRI). Methods: The datasets related to HIRI were sourced from the Gene Expression Omnibus database. The comparative analysis of differential gene expression involving CRGs was performed between HIRI and normal liver samples. Correlation analysis, function enrichment analyses, and protein-protein interactions were employed to understand the interactions and roles of these genes. Machine learning techniques were used to identify hub genes. Additionally, differences in immune cell infiltration between HIRI patients and controls were analyzed. Quantitative real-time PCR and western blotting were used to verify the expression of the hub genes. Results: Seventy-five HIRI and 80 control samples from three databases were included in the bioinformatics analysis. Three hub CRGs (NLRP3, ATP7B and NFE2L2) were identified using three machine learning models. Diagnostic accuracy was assessed using a receiver operating characteristic (ROC) curve for the hub genes, which yielded an area under the ROC curve (AUC) of 0.832. Remarkably, in the validation datasets GSE15480 and GSE228782, the three hub genes had AUC reached 0.904. Additional analyses, including nomograms, decision curves, and calibration curves, supported their predictive power for diagnosis. Enrichment analyses indicated the involvement of these genes in multiple pathways associated with HIRI progression. Comparative assessments using CIBERSORT and gene set enrichment analysis suggested elevated expression of these hub genes in activated dendritic cells, neutrophils, activated CD4 memory T cells, and activated mast cells in HIRI samples versus controls. A ceRNA network underscored a complex regulatory interplay among genes. The genes mRNA and protein levels were also verified in HIRI-affected mouse liver tissues. Conclusion: Our findings have provided a comprehensive understanding of the association between cuproptosis and HIRI, establishing a promising diagnostic pattern and identifying latent therapeutic targets for HIRI treatment. Additionally, our study offers novel insights to delve deeper into the underlying mechanisms of HIRI.


Subject(s)
Computational Biology , Machine Learning , Reperfusion Injury , Humans , Computational Biology/methods , Reperfusion Injury/genetics , Reperfusion Injury/immunology , Reperfusion Injury/diagnosis , Gene Expression Profiling , Liver/metabolism , Liver/immunology , Liver/pathology , Animals , Protein Interaction Maps , Mice , Gene Regulatory Networks , Databases, Genetic , Transcriptome , Male , Biomarkers
8.
Front Immunol ; 15: 1386905, 2024.
Article in English | MEDLINE | ID: mdl-38812509

ABSTRACT

Backgrounds: Non-alcoholic fatty liver disease (NAFLD) presents as a common liver disease characterized by an indistinct pathogenesis. Disulfidptosis is a recently identified mode of cell death. This study aimed to investigate the potential role of disulfidptosis-related genes (DRGs) in the pathogenesis of NAFLD. Methods: Gene expression profiles were obtained from the bulk RNA dataset GSE126848 and the single-cell RNA dataset GSE136103, both associated with NAFLD. Our study assessed the expression of DRGs in NAFLD and normal tissues. Weighted gene co-expression network analysis (WGCNA) and differential expression analysis were employed to identify the key NAFLD-specific differentially expressed DRGs (DE-DRGs). To explore the biological functions and immune regulatory roles of these key DE-DRGs, we conducted immune infiltration analysis, functional enrichment analysis, consensus clustering analysis, and single-cell differential state analysis. Finally, we validated the expression and biological functions of DRGs in NAFLD patients using histology and RNA-sequencing transcriptomic assays with human liver tissue samples. Results: Through the intersection of WGCNA, differentially expressed genes, and DRGs, two key DE-DRGs (DSTN and MYL6) were identified. Immune infiltration analysis indicated a higher proportion of macrophages, T cells, and resting dendritic cells in NAFLD compared to control liver samples. Based on the key DE-DRGs, Two disulfidptosis clusters were defined in GSE126848. Cluster 1, with higher expression of the key DE-DRGs, exhibited increased immune infiltration abundance and was closely associated with oxidative stress and immune regulation compared to cluster 2. High-resolution analysis of mononuclear phagocytes highlighted the potential role of MYL6 in intrahepatic M1 phenotype Kupffer cells in NAFLD patients. Our transcriptome data revealed that the expression levels of the majority of DRGs were significantly increased in NAFLD patients. NAFLD patients exhibit elevated MYL6 correlating with inflammation, oxidative stress, and disease severity, offering promising diagnostic specificity. Conclusion: This comprehensive study provides evidence for the association between NAFLD and disulfidptosis, identifying potential target genes and pathways in NAFLD. The identification of MYL6 as a possible treatment target for NAFLD provided a novel understanding of the disease's development.


Subject(s)
Non-alcoholic Fatty Liver Disease , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/immunology , Non-alcoholic Fatty Liver Disease/pathology , Humans , Gene Expression Profiling , Transcriptome , Gene Regulatory Networks , Liver/metabolism , Liver/pathology , Liver/immunology , Gene Expression Regulation
9.
Front Immunol ; 15: 1376276, 2024.
Article in English | MEDLINE | ID: mdl-38745658

ABSTRACT

Background: Hepatic Inflammatory Pseudotumor (IPT) is an infrequent condition often masquerading as a malignant tumor, resulting in misdiagnosis and unnecessary surgical resection. The emerging concept of IgG4-related diseases (IgG4-RD) has gained widespread recognition, encompassing entities like IgG4-related hepatic IPT. Clinically and radiologically, corticosteroids and immunosuppressive therapies have proven effective in managing this condition. Case Presentation: A 3-year-old Chinese boy presented to the clinic with an 11-month history of anemia, fever of unknown origin, and a tender hepatic mass. Blood examinations revealed chronic anemia (Hb: 6.4 g/L, MCV: 68.6 fl, MCH: 19.5 pg, reticulocytes: 1.7%) accompanied by an inflammatory reaction and an elevated serum IgG4 level (1542.2 mg/L). Abdominal contrast-enhanced computed tomography unveiled a 7.6 cm low-density mass in the right lateral lobe, while magnetic resonance imaging demonstrated slight hypointensity on T1-weighted images and slight hyperintensity on T2-weighted images, prompting suspicion of hepatic malignancy. A subsequent liver biopsy revealed a mass characterized by fibrous stroma and dense lymphoplasmacytic infiltration. Immunohistochemical analysis confirmed the presence of IgG4-positive plasma cells, leading to the diagnosis of IgG4-related hepatic IPT. Swift resolution occurred upon initiation of corticosteroid and mycophenolate mofetil therapies. Conclusion: This study underscores the diagnostic approach to hepatic IPT, utilizing histopathology, immunostaining, imaging, serology, organ involvement, and therapeutic response. Early histological examination plays a pivotal role in clinical guidance, averting misdiagnosis as a liver tumor and unnecessary surgical interventions.


Subject(s)
Granuloma, Plasma Cell , Immunoglobulin G4-Related Disease , Immunoglobulin G , Humans , Male , Granuloma, Plasma Cell/diagnosis , Granuloma, Plasma Cell/immunology , Granuloma, Plasma Cell/drug therapy , Child, Preschool , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin G4-Related Disease/diagnosis , Liver Diseases/diagnosis , Liver Diseases/immunology , Diagnosis, Differential , Liver/pathology , Liver/diagnostic imaging , Liver/immunology , Tomography, X-Ray Computed , Biopsy , Immunosuppressive Agents/therapeutic use
10.
Front Immunol ; 15: 1400553, 2024.
Article in English | MEDLINE | ID: mdl-38817615

ABSTRACT

Background and aims: Metabolic dysfunction-associated steatohepatitis (MASH) is a significant health concern with limited treatment options. AXL, a receptor tyrosine kinase activated by the GAS6 ligand, promotes MASH through activation of hepatic stellate cells and inflammatory macrophages. This study identified cell subsets affected by MASH progression and the effect of AXL inhibition. Methods: Mice were fed chow or different fat-enriched diets to induce MASH, and small molecule AXL kinase inhibition with bemcentinib was evaluated. Gene expression was measured by qPCR. Time-of-flight mass cytometry (CyTOF) used single cells from dissociated livers, acquired on the Fluidigm Helios, and cell populations were studied using machine learning. Results: In mice fed different fat-enriched diets, liver steatosis alone was insufficient to elevate plasma soluble AXL (sAXL) levels. However, in conjunction with inflammation, sAXL increases, serving as an early indicator of steatohepatitis progression. Bemcentinib, an AXL inhibitor, effectively reduced proinflammatory responses in MASH models, even before fibrosis appearance. Utilizing CyTOF analysis, we detected a decreased population of Kupffer cells during MASH while promoting infiltration of monocytes/macrophages and CD8+ T cells. Bemcentinib partially restored Kupffer cells, reduced pDCs and GzmB- NK cells, and increased GzmB+CD8+ T cells and LSECs. Additionally, AXL inhibition enhanced a subtype of GzmB+CD8+ tissue-resident memory T cells characterized by CX3CR1 expression. Furthermore, bemcentinib altered the transcriptomic landscape associated with MASH progression, particularly in TLR signaling and inflammatory response, exhibiting differential cytokine expression in the plasma, consistent with liver repair and decreased inflammation. Conclusion: Our findings highlight sAXL as a biomarker for monitoring MASH progression and demonstrate that AXL targeting shifted liver macrophages and CD8+ T-cell subsets away from an inflammatory phenotype toward fibrotic resolution and organ healing, presenting a promising strategy for MASH treatment.


Subject(s)
Axl Receptor Tyrosine Kinase , Liver Cirrhosis , Proto-Oncogene Proteins , Receptor Protein-Tyrosine Kinases , Animals , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/metabolism , Mice , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Liver Cirrhosis/immunology , Liver Cirrhosis/drug therapy , Liver Cirrhosis/pathology , Liver Cirrhosis/metabolism , Male , Disease Models, Animal , Mice, Inbred C57BL , Benzocycloheptenes/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Liver/pathology , Liver/immunology , Liver/metabolism , Liver/drug effects , Macrophages/immunology , Macrophages/metabolism , Macrophages/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Triazoles
11.
Viruses ; 16(5)2024 05 07.
Article in English | MEDLINE | ID: mdl-38793619

ABSTRACT

BACKGROUND AND AIMS: The outcomes of HBV infections are related to complex immune imbalances; however, the precise mechanisms by which HBV induces immune dysfunction are not well understood. METHODS: HBV transgenic (HBs-Tg) mice were used to investigate intrahepatic NK cells in two distinct subsets: conventional NK (cNK) and liver-resident NK (LrNK) cells during a chronic HBV infection. RESULTS: The cNK cells, but not the LrNK cells, were primarily responsible for the increase in the number of bulk NK cells in the livers of ageing HBs-Tg mice. The hepatic cNK cells showed a stronger ability to produce IL-10, coupled with a higher expression of CD69, TIGIT and PD-L1, and lower NKG2D expression in ageing HBs-Tg mice. A lower mitochondrial mass and membrane potential, and less polarized localization were observed in the hepatic cNK cells compared with the splenic cNK cells in the HBs-Tg mice. The enhanced galectin-3 (Gal-3) secreted from HBsAg+ hepatocytes accounted for the IL-10 production of hepatic cNK cells via ITGB1 signaling. For humans, LGALS3 and ITGB1 expression is positively correlated with IL-10 expression, and negatively correlated with the poor clinical progression of HCC. CONCLUSIONS: Gal-3-ITGB1 signaling shapes hepatic cNK cells but not LrNK cells during a chronic HBV infection, which may correlate with HCC progression.


Subject(s)
Carcinoma, Hepatocellular , Galectin 3 , Hepatitis B virus , Interleukin-10 , Killer Cells, Natural , Liver Neoplasms , Liver , Mice, Transgenic , Signal Transduction , Animals , Mice , Killer Cells, Natural/immunology , Humans , Hepatitis B virus/genetics , Hepatitis B virus/immunology , Interleukin-10/genetics , Interleukin-10/metabolism , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/virology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/immunology , Liver Neoplasms/virology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver/pathology , Liver/immunology , Liver/virology , Liver/metabolism , Galectin 3/genetics , Galectin 3/metabolism , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/virology , Disease Progression , Male , Female , Hepatocytes/virology , Hepatocytes/metabolism , Hepatocytes/immunology , Mice, Inbred C57BL , Galectins/genetics , Galectins/metabolism
12.
Trends Parasitol ; 40(6): 446-448, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38772757

ABSTRACT

Liebold et al. recently revealed how the identity of dying cells drives distinct changes to the macrophages which engulf and clear them, a process known as efferocytosis. During infection with the helminth Schistosoma mansoni, liver macrophages recapitulate these phenotypes, mediated by Axl/MerTK receptors and regulating egg burdens.


Subject(s)
Macrophages , Phagocytosis , Schistosoma mansoni , Animals , Macrophages/immunology , Macrophages/parasitology , Schistosoma mansoni/physiology , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/parasitology , Receptor Protein-Tyrosine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/immunology , Humans , Liver/parasitology , Liver/immunology , Axl Receptor Tyrosine Kinase , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , c-Mer Tyrosine Kinase/metabolism , c-Mer Tyrosine Kinase/physiology , Efferocytosis
13.
Int Immunopharmacol ; 135: 112277, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38788445

ABSTRACT

Sepsis, a systemic inflammatory response triggered by infection, has a considerably high mortality rate. However, effective prevention and intervention measures against sepsis remain insufficient. Therefore, this study aimed to investigate the mechanisms underlying the protective properties of immune response gene-1 (IRG1) and 4-Octyl itaconate (OI) during acute liver damage in mice with sepsis. A sepsis mouse model was established to compare wild-type and IRG1-/- groups. The impact of IRG1/Itaconate on pro- and anti-inflammatory cytokines was evaluated using J774A.1 cells. IRG1/Itaconate substantially reduced pro-inflammatory cytokines and increased the release of anti-inflammatory cytokines. It reduced pathological damage to liver tissues, preserved normal liver function, decreased the release of reactive oxygen species (ROS) and LDH, and enhanced the GSH/GSSG ratio. Moreover, IRG1 and itaconic acid activated the Nrf2 signaling pathway, regulating the expression of its downstream antioxidative stress-related proteins. Additionally, they inhibited the activity of NLRP3 inflammatory vesicles to suppress the expression of macrophage-associated pyroptosis signaling molecules. Our findings demonstrate that IRG1/OI inhibits NLRP3 inflammatory vesicle activation and macrophage pyroptosis by modulating the Nrf2 signaling pathway, thereby attenuating acute liver injury in mice with sepsis. These findings could facilitate the clinical application of IRG1/Itaconate to prevent sepsis-induced acute liver injury.


Subject(s)
Macrophages , Mice, Inbred C57BL , NF-E2-Related Factor 2 , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Sepsis , Signal Transduction , Succinates , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Succinates/therapeutic use , Succinates/pharmacology , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Sepsis/drug therapy , Sepsis/complications , Sepsis/immunology , Pyroptosis/drug effects , Mice , Signal Transduction/drug effects , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Male , Mice, Knockout , Liver/pathology , Liver/drug effects , Liver/metabolism , Liver/immunology , Cell Line , Disease Models, Animal , Cytokines/metabolism , Hydro-Lyases/metabolism , Reactive Oxygen Species/metabolism , Humans , Carboxy-Lyases/metabolism , Carboxy-Lyases/genetics
14.
Nat Rev Immunol ; 24(6): 379, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38719946
15.
Sci Adv ; 10(22): eadk5011, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38809975

ABSTRACT

Healthy behavioral patterns could modulate organ functions to enhance the body's immunity. However, how exercise regulates antiviral innate immunity remains elusive. Here, we found that exercise promotes type I interferon (IFN-I) production in the liver and enhances IFN-I immune activity of the body. Despite the possibility that many exercise-induced factors could affect IFN-I production, we identified Gpld1 as a crucial molecule, and the liver as the major organ to promote IFN-I production after exercise. Exercise largely loses the efficiency to induce IFN-I in Gpld1-/- mice. Further studies demonstrated that exercise-produced 3-hydroxybutanoic acid (3-HB) critically induces Gpld1 expression in the liver. Gpld1 blocks the PP2A-IRF3 interaction, thus enhancing IRF3 activation and IFN-I production, and eventually improving the body's antiviral ability. This study reveals that exercise improves antiviral innate immunity by linking the liver metabolism to systemic IFN-I activity and uncovers an unknown function of liver cells in innate immunity.


Subject(s)
Immunity, Innate , Interferon Regulatory Factor-3 , Interferon Type I , Liver , Physical Conditioning, Animal , Animals , Male , Mice , Antiviral Agents , Cytokines , Interferon Regulatory Factor-3/metabolism , Interferon Type I/metabolism , Liver/metabolism , Liver/immunology , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Ubiquitins , Glycosylphosphatidylinositol Diacylglycerol-Lyase/metabolism
16.
Nat Immunol ; 25(6): 981-993, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38811816

ABSTRACT

Viral infection makes us feel sick as the immune system alters systemic metabolism to better fight the pathogen. The extent of these changes is relative to the severity of disease. Whether blood glucose is subject to infection-induced modulation is mostly unknown. Here we show that strong, nonlethal infection restricts systemic glucose availability, which promotes the antiviral type I interferon (IFN-I) response. Following viral infection, we find that IFNγ produced by γδ T cells stimulates pancreatic ß cells to increase glucose-induced insulin release. Subsequently, hyperinsulinemia lessens hepatic glucose output. Glucose restriction enhances IFN-I production by curtailing lactate-mediated inhibition of IRF3 and NF-κB signaling. Induced hyperglycemia constrained IFN-I production and increased mortality upon infection. Our findings identify glucose restriction as a physiological mechanism to bring the body into a heightened state of responsiveness to viral pathogens. This immune-endocrine circuit is disrupted in hyperglycemia, possibly explaining why patients with diabetes are more susceptible to viral infection.


Subject(s)
Blood Glucose , Immunity, Innate , Interferon-gamma , Animals , Interferon-gamma/metabolism , Interferon-gamma/immunology , Mice , Blood Glucose/metabolism , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/metabolism , Mice, Inbred C57BL , Signal Transduction/immunology , Insulin/metabolism , Insulin/immunology , Mice, Knockout , Hyperglycemia/immunology , Interferon Regulatory Factor-3/metabolism , NF-kappa B/metabolism , Humans , Liver/immunology , Liver/virology , Liver/metabolism , Male
17.
Front Immunol ; 15: 1379225, 2024.
Article in English | MEDLINE | ID: mdl-38650949

ABSTRACT

Dendritic cells (DCs) are major antigen-presenting cells that connect innate and adaptive immunity. Hepatic DCs are less activated and contribute to maintain the tolerogenic environment of the liver under steady state. Several studies indicated DCs in metabolic dysfunction-associated steatohepatitis (MASH), representing a substantial burden on healthcare systems due to its association with liver-related morbidity and mortality. Studies highlighted the potential disease-promoting role of liver DCs in the development of MASH while other experimental systems suggested their protective role. This review discusses this controversy and the current understanding of how DCs affect the pathogenesis of MASH.


Subject(s)
Dendritic Cells , Dendritic Cells/immunology , Humans , Animals , Liver/immunology , Fatty Liver/immunology
18.
Vet Res ; 55(1): 53, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38658996

ABSTRACT

Gene expression for Th1/Th2 cytokines (IL-4 and IFN-É£), regulatory cytokines (TGF-ß and IL-10) and the transcriptional factor FoxP3 was analyzed in the liver and hepatic lymph nodes (HLN) from sheep immunized with partially protective and non-protective vaccine candidates and challenged with Fasciola hepatica. FoxP3 T cells were also evaluated by immunohistochemistry (IHQ). The most remarkable difference between the partially protected vaccinated (V1) group and the non-protected vaccinated (V2) group was a more severe expansion of FoxP3 T cells recorded by IHQ in both the liver and HLN of the V2 group as compared to the V1 group, whereas no differences were found between the V2 group and the infected control (IC) group. Similar results were recorded for FoxP3 gene expression although significant differences among V1 and V2 groups were only significant in the HLN, while FoxP3 gene expression was very similar in the V2 and IC groups both in the liver and HLN. No significant differences for the remaining cytokines were recorded between the V1 and V2 groups, but in the liver the V2 group shows significant increases of IFN-É£ and IL-10 as compared to the uninfected control (UC) group whereas the V1 group did not. The lower expansion of FoxP3 T cells and lower increase of IFN-É£ and IL-10 in the partially protected vaccinated group may be related with lower hepatic lesions and fluke burdens recorded in this group as compared to the other two infected groups. The most relevant change in regulatory cytokine gene expression was the significant increase of TGF-ß in the liver of IC, V1 and V2 groups as compared to the UC group, which could be related to hepatic lesions.


Subject(s)
Cytokines , Fasciola hepatica , Fascioliasis , Forkhead Transcription Factors , Sheep Diseases , Animals , Fascioliasis/veterinary , Fascioliasis/prevention & control , Fascioliasis/immunology , Fasciola hepatica/immunology , Sheep , Forkhead Transcription Factors/metabolism , Sheep Diseases/prevention & control , Sheep Diseases/immunology , Sheep Diseases/parasitology , Cytokines/metabolism , Liver/parasitology , Liver/immunology , Vaccines/immunology , Vaccines/administration & dosage , Th1 Cells/immunology , Lymph Nodes/immunology , Female , Th2 Cells/immunology
19.
Acta Trop ; 254: 107208, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38621620

ABSTRACT

The study aimed to elicit protective immune responses against murine schistosomiasis mansoni at the parasite lung- and liver stage. Two peptides showing amino acid sequence similarity to gut cysteine peptidases, which induce strong memory immune effectors in the liver, were combined with a peptide based on S. mansoni thioredoxin peroxidase (TPX), a prominent lung-stage schistosomula excretory-secretory product, and alum as adjuvant. Only one of the 2 cysteine peptidases-based peptides in a multiple antigenic peptide construct (MAP-3 and MAP-4) appeared to adjuvant protective immune responses induced by the TPX peptide in a MAP form. Production of TPX MAP-specific IgG1 serum antibodies, and increase in lung interleukin-1 (IL-1), uric acid, and reactive oxygen species (ROS) content were associated with significant (P < 0.05) 50 % reduction in recovery of lung-stage larvae. Increase in lung triglycerides and cholesterol levels appeared to provide the surviving worms with nutrients necessary for a stout double lipid bilayer barrier at the parasite-host interface. Surviving worms-released products elicited memory responses to the MAP-3 immunogen, including production of specific IgG1 antibodies and increase in liver IL-33 and ROS. Reduction in challenge worm burden recorded 45 days post infection did not exceed 48 % associated with no differences in parasite egg counts in the host liver and small intestine compared to unimmunized adjuvant control mice. Alum adjuvant assisted the second peptide, MAP-4, in production of IgG1, IgG2a, IgG2b and IgA specific antibodies and increase in liver ROS, but with no protective potential, raising doubt about the necessity of adjuvant addition. Accordingly, different vaccine formulas containing TPX MAP and 1, 2 or 3 cysteine peptidases-derived peptides with or without alum were used to immunize parallel groups of mice. Compared to unimmunized control mice, significant (P < 0.05 to < 0.005) 22 to 54 % reduction in worm burden was recorded in the different groups associated with insignificant changes in parasite egg output. The results together indicated that a schistosomiasis vaccine able to entirely prevent disease and halt its transmission still remains elusive.


Subject(s)
Adjuvants, Immunologic , Antibodies, Helminth , Immunoglobulin G , Liver , Lung , Schistosoma mansoni , Schistosomiasis mansoni , Vaccines, Subunit , Animals , Schistosoma mansoni/immunology , Schistosomiasis mansoni/prevention & control , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/parasitology , Lung/parasitology , Lung/immunology , Mice , Antibodies, Helminth/immunology , Antibodies, Helminth/blood , Liver/parasitology , Liver/immunology , Immunoglobulin G/blood , Adjuvants, Immunologic/administration & dosage , Vaccines, Subunit/immunology , Vaccines, Subunit/administration & dosage , Female , Antigens, Helminth/immunology , Disease Models, Animal , Alum Compounds/administration & dosage , Mice, Inbred BALB C , Protein Subunit Vaccines
20.
J Med Virol ; 96(5): e29627, 2024 May.
Article in English | MEDLINE | ID: mdl-38659381

ABSTRACT

The immune mechanism underlying hepatitis B surface antigen (HBsAg) loss, particularly type I inflammatory response, during pegylated interferon-α (PEG-IFN) therapy remains unclear. In this study, we aimed to elucidate such immune mechanisms. Overall, 82 patients with chronic hepatitis B (CHB), including 41 with HBsAg loss (cured group) and 41 uncured patients, received nucleos(t)ide analogue and PEG-IFN treatments. Blood samples from all patients, liver tissues from 14 patients with CHB, and hepatic perfusate from 8 liver donors were collected for immune analysis. Jurkat, THP-1 and HepG2.2.15 cell lines were used in cell experiments. The proportion of IFN-γ+ Th1 cells was higher in the cured group than in the uncured group, which was linearly correlated with HBsAg decline and alanine aminotransferase (ALT) levels during treatment. However, CD8+ T cells were weakly associated with HBsAg loss. Serum and intrahepatic levels of Th1 cell-associated chemokines (C-X-C motif chemokine ligand [CXCL] 9, CXCL10, CXCL11, IFN-γ) were significantly lower in the cured patients than in patients with a higher HBsAg quantification during therapy. Serum from cured patients induced more M1 (CD68+CD86+ macrophage) cells than that from uncured patients. Patients with chronic HBV infection had significantly lower proportions of CD86+ M1 and CD206+ M2 macrophages in their livers than healthy controls. M1 polarization of intrahepatic Kupffer cells promoted HBsAg loss by upregulating the effector function of tissue-resident memory T cells with increased ALT levels. IFN-γ+ Th1 activates intrahepatic resident memory T cells to promote HBsAg loss by inducing M1 macrophage polarization.


Subject(s)
Hepatitis B Surface Antigens , Hepatitis B, Chronic , Liver , Macrophages , Memory T Cells , Th1 Cells , Adult , Female , Humans , Male , Middle Aged , Antiviral Agents/therapeutic use , Antiviral Agents/pharmacology , Hepatitis B Surface Antigens/immunology , Hepatitis B virus/immunology , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/drug therapy , Interferon-alpha , Interferon-gamma , Liver/immunology , Macrophages/immunology , Memory T Cells/immunology , Th1 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...