Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 401
Filter
1.
Cell Rep Med ; 5(5): 101564, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38733988

ABSTRACT

Monocytes (Mos) are crucial in the evolution of metabolic dysfunction-associated steatotic liver disease (MASLD) to metabolic dysfunction-associated steatohepatitis (MASH), and immunometabolism studies have recently suggested targeting leukocyte bioenergetics in inflammatory diseases. Here, we reveal a peculiar bioenergetic phenotype in circulating Mos of patients with MASH, characterized by high levels of glycolysis and mitochondrial (mt) respiration. The enhancement of mt respiratory chain activity, especially complex II (succinate dehydrogenase [SDH]), is unbalanced toward the production of reactive oxygen species (ROS) and is sustained at the transcriptional level with the involvement of the AMPK-mTOR-PGC-1α axis. The modulation of mt activity with dimethyl malonate (DMM), an SDH inhibitor, restores the metabolic profile and almost abrogates cytokine production. Analysis of a public single-cell RNA sequencing (scRNA-seq) dataset confirms that in murine models of MASH, liver Mo-derived macrophages exhibit an upregulation of mt and glycolytic energy pathways. Accordingly, the DMM injection in MASH mice contrasts Mo infiltration and macrophagic enrichment, suggesting immunometabolism as a potential target in MASH.


Subject(s)
Energy Metabolism , Mitochondria , Monocytes , Humans , Animals , Monocytes/metabolism , Monocytes/immunology , Mice , Mitochondria/metabolism , Fatty Liver/metabolism , Fatty Liver/pathology , Fatty Liver/immunology , Male , Glycolysis , Reactive Oxygen Species/metabolism , Mice, Inbred C57BL , Macrophages/metabolism , Macrophages/immunology , Female , Liver/metabolism , Liver/pathology
2.
Int Immunopharmacol ; 133: 112091, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38657500

ABSTRACT

The pathophysiology of hepatic steatosis is thoroughly reviewed in this comprehensive report, with particular attention to the complex interactions between inflammatory pathways, insulin resistance, lipid metabolism, metabolic dysregulation, and immunological responses in the liver including non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), and hepatocellular carcinoma (HCC). The study highlights the role of immune cell regulation in disease progression and explores the potential of immune cell-specific treatments for treating hepatic disorders. The development of liver disorders is significantly influenced by immune cells, including dendritic cells, T cells, and natural killer cells. Clinical investigations show that immune cell-specific treatments can effectively reduce liver fibrosis and inflammation. Future research should focus on finding new immunological targets for therapeutic interventions, as well as addressing the management challenges associated with NAFLD/NASH. Hepatic immune microorganisms also impact liver homeostasis and disorders. Improvements in immune cell regulation and liver transplantation methods give patients hope for better prognoses. Important phases include optimizing the selection of donors for malignancy of the liver, using machine perfusion for organ preservation, and fine-tuning immunosuppressive strategies. For focused treatments in hepatic steatosis, it is imperative to understand the intricate interactions between immune and metabolic variables. Understanding the liver's heterogeneous immune profile, encompassing a range of immune cell subpopulations, is crucial for formulating focused therapeutic interventions. To improve patient care and outcomes in hepatic illnesses, there is an urgent need for further research and innovation. Therefore, to effectively treat hepatic steatosis, it is important to enhance therapeutic techniques and maximize liver transplantation strategies.


Subject(s)
Non-alcoholic Fatty Liver Disease , Humans , Animals , Non-alcoholic Fatty Liver Disease/immunology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/therapy , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/therapy , Liver Neoplasms/immunology , Liver Neoplasms/metabolism , Liver Neoplasms/therapy , Liver/immunology , Liver/metabolism , Liver/pathology , Lipid Metabolism , Insulin Resistance/immunology , Fatty Liver/immunology , Fatty Liver/metabolism , Fatty Liver/therapy
3.
Biomolecules ; 14(4)2024 Apr 13.
Article in English | MEDLINE | ID: mdl-38672492

ABSTRACT

Metabolic dysfunction-associated steatotic liver disease (MASLD) has emerged as the most common liver disease worldwide in recent years. MASLD commonly presents as simple hepatic steatosis, but ~25% of patients develop liver inflammation, progressive fibrosis, liver cirrhosis and related hepatocellular carcinoma. Liver inflammation and the degree of fibrosis are key determinants of the prognosis. The pathophysiology of liver inflammation is incompletely understood and involves diverse factors and specifically innate and adaptive immune responses. More specifically, diverse mediators of innate immunity such as proinflammatory cytokines, adipokines, inflammasomes and various cell types like mononuclear cells, macrophages and natural killer cells are involved in directing the inflammatory process in MASLD. The activation of innate immunity is driven by various factors including excess lipids and lipotoxicity, insulin resistance and molecular patterns derived from gut commensals. Targeting pathways of innate immunity might therefore appear as an attractive therapeutic strategy in the future management of MASLD and possibly its complications.


Subject(s)
Immunity, Innate , Humans , Animals , Fatty Liver/immunology , Inflammasomes/immunology , Inflammasomes/metabolism , Cytokines/metabolism , Cytokines/immunology , Insulin Resistance/immunology , Inflammation/immunology
4.
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
5.
J Nutr Biochem ; 99: 108852, 2022 01.
Article in English | MEDLINE | ID: mdl-34525389

ABSTRACT

This study aimed to develop a well-characterized mouse model of alcoholic hepatitis (AH) regression. Male C57BL/6J mice were fed a Lieber-DeCarli (LD) control diet or LD containing 5% ethanol for ten days followed by one binge, which is the chronic-binge model of AH developed by the National Institute on Alcohol Abuse and Alcoholism. To determine AH regression, mice previously exposed to ethanol were put on LD control diet and metabolic and inflammatory features were monitored weekly for three weeks. Serum alcohol, total cholesterol, and alanine transaminase levels were increased in ethanol-fed mice, which declined to those of no ethanol controls within one and three weeks after ethanol withdrawal, respectively. Serum malondialdehyde was increased with ethanol feeding, but it was restored to no ethanol control levels within one week. Ethanol-induced changes in the hepatic expression of genes involved in lipogenesis, fatty acid oxidation, ethanol metabolism, and antioxidant response were restored to those of no ethanol controls after 3 weeks of ethanol withdrawal. Also, ethanol-induced hepatic inflammation was gradually decreased during the 3 weeks of ethanol withdrawal. Hepatic nicotinamide adenine dinucleotide (NAD+) levels and the expression of enzymes involved in the NAD+ salvage pathway were decreased by ethanol feeding, which was mitigated after ethanol withdrawal. Ethanol significantly lowered hepatic sirtuin 1 expression, but its levels were restored with ethanol cessation. This study established a mouse model of AH regression, which can be used as a preclinical model to study the potential of dietary bioactives or therapeutic agents on AH regression.


Subject(s)
Alcoholism/complications , Ethanol/adverse effects , Fatty Liver/metabolism , Hepatitis, Alcoholic/metabolism , NAD/metabolism , Oxidative Stress , Animals , Disease Models, Animal , Disease Progression , Fatty Liver/etiology , Fatty Liver/genetics , Fatty Liver/immunology , Hepatitis, Alcoholic/etiology , Hepatitis, Alcoholic/genetics , Hepatitis, Alcoholic/immunology , Humans , Liver/immunology , Liver/metabolism , Male , Malondialdehyde/metabolism , Mice , Mice, Inbred C57BL , Sirtuin 1/genetics , Sirtuin 1/metabolism
6.
Int J Mol Sci ; 22(23)2021 Nov 30.
Article in English | MEDLINE | ID: mdl-34884762

ABSTRACT

Immune checkpoint inhibitors represent one of the most significant recent advances in clinical oncology, since they dramatically improved the prognosis of deadly cancers such as melanomas and lung cancer. Treatment with these drugs may be complicated by the occurrence of clinically-relevant adverse drug reactions, most of which are immune-mediated, such as pneumonitis, colitis, endocrinopathies, nephritis, Stevens Johnson syndrome and toxic epidermal necrolysis. Drug-induced steatosis and steatohepatitis are not included among the typical forms of cancer immunotherapy-induced liver toxicity, which, instead, usually occurs as a panlobular hepatitis with prominent lymphocytic infiltrates. Nonetheless, non-alcoholic fatty liver disease is a risk factor for immunotherapy-induced hepatitis, and steatosis and steatohepatitis are frequently observed in this condition. In the present review we discuss how these pathology findings could be explained in the context of current models suggesting immune-mediated pathogenesis for steatohepatitis. We also review evidence suggesting that in patients with hepatocellular carcinoma, the presence of steatosis or steatohepatitis could predict a poor therapeutic response to these agents. How these findings could fit with immune-mediated mechanisms of these liver diseases will also be discussed.


Subject(s)
Fatty Liver/etiology , Immune Checkpoint Inhibitors/adverse effects , Immunotherapy/adverse effects , Neoplasms/therapy , Carcinoma, Hepatocellular/complications , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/therapy , Fatty Liver/immunology , Humans , Liver/drug effects , Liver/pathology , Liver Neoplasms/complications , Liver Neoplasms/immunology , Liver Neoplasms/therapy , Models, Biological , Neoplasms/immunology , Non-alcoholic Fatty Liver Disease/complications , Prognosis , Risk Factors
7.
Front Immunol ; 12: 726829, 2021.
Article in English | MEDLINE | ID: mdl-34691032

ABSTRACT

ß2 integrins are critical for neutrophil firm adhesion, trans-endothelial migration, and the recruitment to the inflamed tissue. Autophagy is implicated in cell migration and tumor metastasis through facilitating the turnover of ß1 integrins; however, whether autophagy is able to control neutrophil migration by promoting the degradation of ß2 integrins is unexplored. Here, we show that high blood levels of palmitic acid (PA) strongly triggered neutrophil autophagy activation, leading to adhesion deficiency in dairy cows with fatty liver. The three neutrophil granule subtypes, namely, azurophil granules (AGs), specific granules (SGs), and gelatinase granules (GGs), were engulfed by the autophagosomes for degradation, resulting in an increased vacuolation in fatty liver dairy cow neutrophils. Importantly, the adhesion-associated molecules CD11b and CD18 distributed on AGs, SGs, and GGs were degraded with the three granule subtypes by autophagy. Moreover, FGA, Hsc70, and TRIM21 mediated the degradation of cytosolic oxidized-ubiquitinated CD11b and CD18. Collectively, our results demonstrate that high blood PA triggers neutrophil autophagy-dependent vacuolation and granule-dependent adhesion deficiency, decreasing neutrophil mobility, and impairing the innate immune system of dairy cow with fatty liver. This theory extends the category of autophagy in maintaining granule homeostasis and provides a novel strategy to improve the immune of dairy cows with metabolic disease.


Subject(s)
Autophagy , Cell Adhesion , Fatty Liver/immunology , Neutrophils/physiology , Palmitic Acid/blood , Animals , Autophagy-Related Protein 5/genetics , CD11b Antigen/immunology , CD18 Antigens/immunology , Cattle , Fatty Liver/blood , Female , Fibrinogen/genetics , HL-60 Cells , HSC70 Heat-Shock Proteins/genetics , Humans , Macrophage-1 Antigen , Ribonucleoproteins/genetics
8.
Cell Prolif ; 54(10): e13116, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34469018

ABSTRACT

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.


Subject(s)
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
9.
Front Immunol ; 12: 694720, 2021.
Article in English | MEDLINE | ID: mdl-34248992

ABSTRACT

The present study aimed to elucidate the mechanism of dietary betaine, as a lipid-lowering substance, on the regulation of lipid metabolism and inflammation in juvenile black seabream (Acanthopagrus schlegelii) fed a high fat diet. An 8-week feeding trial was conducted in black seabream with an initial weight of 8.39 ± 0.01g fed four isonitrogenous diets including Control, medium-fat diet (11%); HFD, high-fat diet (17%); and HFD supplemented with two levels (10 and 20 g/kg) of betaine, HFD+B1 and HFD+B2, respectively. SGR and FE in fish fed HFD+B2 were significantly higher than in fish fed HFD. Liver histology revealed that vacuolar fat droplets were smaller and fewer in bream fed HFD supplemented with betaine compared to fish fed HFD. Betaine promoted the mRNA and protein expression levels of silent information regulator 1 (Sirt1), up-regulated mRNA expression and protein content of lipid peroxisome proliferator-activated receptor alpha (pparα), and down-regulated mRNA expression and protein content of sterol regulatory element-binding protein-1(srebp-1). Furthermore, the mRNA expression levels of anti-inflammatory cytokines in liver and intestine were up-regulated, while nuclear factor kB (nf-kb) and pro-inflammatory cytokines were down-regulated by dietary betaine supplementation. Likewise, in fish that received lipopolysaccharide (LPS) to stimulate inflammatory responses, the expression levels of mRNAs of anti-inflammatory cytokines in liver, intestine and kidney were up-regulated in fish fed HFD supplemented with betaine compared with fish fed HFD, while nf-kb and pro-inflammatory cytokines were down-regulated. This is the first report to suggest that dietary betaine could be an effective feed additive to alleviate hepatic steatosis and attenuate inflammatory responses in black seabream fed a high fat diet by modulating the Sirt1/Srebp-1/Pparɑ pathway.


Subject(s)
Betaine/administration & dosage , Diet, High-Fat/adverse effects , Dietary Supplements , Fatty Liver/veterinary , Fish Diseases/prevention & control , Fish Proteins/metabolism , Inflammation/veterinary , Liver/enzymology , PPAR alpha/metabolism , Sea Bream/metabolism , Sirtuin 1/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Age Factors , Animal Feed , Animals , Cytokines/genetics , Cytokines/metabolism , Fatty Liver/enzymology , Fatty Liver/immunology , Fatty Liver/prevention & control , Fish Diseases/enzymology , Fish Diseases/immunology , Fish Proteins/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation/prevention & control , Liver/immunology , PPAR alpha/genetics , Sea Bream/genetics , Sea Bream/immunology , Sirtuin 1/genetics , Sterol Regulatory Element Binding Protein 1/genetics
10.
Sci Rep ; 11(1): 12314, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34112902

ABSTRACT

We tested the hypothesis that a particular immune activation profile might be correlated with insulin resistance in a general population. By measuring 43 markers of immune, endothelial, and coagulation activation, we have previously shown that five different immune activation profiles may be distinguished in 150 volunteers. One of these profiles, Profile 2, characterized by CD4+ T cell senescence, inflammation, monocyte, B cell, and endothelial activation, presented elevated insulinemia, glycemia, triglyceridemia, and γ-glutamyl transferase, a marker of liver injury, in comparison with other profiles. Our data are compatible with a model in which a particular immune activation profile might favor the development of insulin resistance and metabolic syndrome. In this hypothesis, identification of this profile, that is feasible with only 3 markers with an error rate of 5%, might allow to personalize the screening and prevention of metabolic syndrome-driven morbidities as liver steatosis.


Subject(s)
Inflammation/immunology , Insulin Resistance/immunology , Metabolic Syndrome/immunology , T-Lymphocytes/immunology , gamma-Glutamyltransferase/genetics , Aged , B-Lymphocytes/immunology , Biomarkers/blood , Blood Glucose , CD4-Positive T-Lymphocytes/immunology , Cellular Senescence/genetics , Fatty Liver/genetics , Fatty Liver/immunology , Female , Humans , Inflammation/genetics , Inflammation/pathology , Insulin Resistance/genetics , Male , Metabolic Syndrome/genetics , Metabolic Syndrome/pathology , Middle Aged , Monocytes/immunology , T-Lymphocytes/pathology
11.
Nat Med ; 27(6): 1043-1054, 2021 06.
Article in English | MEDLINE | ID: mdl-34017133

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) are prevalent liver conditions that underlie the development of life-threatening cirrhosis, liver failure and liver cancer. Chronic necro-inflammation is a critical factor in development of NASH, yet the cellular and molecular mechanisms of immune dysregulation in this disease are poorly understood. Here, using single-cell transcriptomic analysis, we comprehensively profiled the immune composition of the mouse liver during NASH. We identified a significant pathology-associated increase in hepatic conventional dendritic cells (cDCs) and further defined their source as NASH-induced boost in cycling of cDC progenitors in the bone marrow. Analysis of blood and liver from patients on the NAFLD/NASH spectrum showed that type 1 cDCs (cDC1) were more abundant and activated in disease. Sequencing of physically interacting cDC-T cell pairs from liver-draining lymph nodes revealed that cDCs in NASH promote inflammatory T cell reprogramming, previously associated with NASH worsening. Finally, depletion of cDC1 in XCR1DTA mice or using anti-XCL1-blocking antibody attenuated liver pathology in NASH mouse models. Overall, our study provides a comprehensive characterization of cDC biology in NASH and identifies XCR1+ cDC1 as an important driver of liver pathology.


Subject(s)
Dendritic Cells/immunology , Fatty Liver/immunology , Non-alcoholic Fatty Liver Disease/immunology , Receptors, Chemokine/genetics , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Cellular Reprogramming/genetics , Cellular Reprogramming/immunology , Dendritic Cells/pathology , Diet, High-Fat/adverse effects , Disease Models, Animal , Fatty Liver/genetics , Fatty Liver/pathology , Female , Humans , Liver/immunology , Liver/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Male , Mice , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Receptors, Chemokine/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
12.
Front Immunol ; 12: 651728, 2021.
Article in English | MEDLINE | ID: mdl-33859644

ABSTRACT

The coronavirus infectious disease 2019 (COVID-19) pandemic has hit the world, affecting health, medical care, economies and our society as a whole. Furthermore, COVID-19 pandemic joins the increasing prevalence of metabolic syndrome in western countries. Patients suffering from obesity, type II diabetes mellitus, cardiac involvement and metabolic associated fatty liver disease (MAFLD) have enhanced risk of suffering severe COVID-19 and mortality. Importantly, up to 25% of the population in western countries is susceptible of suffering from both MAFLD and COVID-19, while none approved treatment is currently available for any of them. Moreover, it is well known that exacerbated innate immune responses are key in the development of the most severe stages of MAFLD and COVID-19. In this review, we focus on the role of the immune system in the establishment and progression of MAFLD and discuss its potential implication in the development of severe COVID-19 in MAFLD patients. As a result, we hope to clarify their common pathology, but also uncover new potential therapeutic targets and prognostic biomarkers for further research.


Subject(s)
Adaptive Immunity/immunology , COVID-19/immunology , COVID-19/pathology , Fatty Liver/immunology , Immunity, Innate/immunology , Angiotensin-Converting Enzyme 2/immunology , Angiotensin-Converting Enzyme 2/metabolism , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 2/pathology , Fatty Liver/pathology , Humans , Liver/immunology , Liver/pathology , Obesity/pathology , Risk Factors , SARS-CoV-2/immunology , Severity of Illness Index
13.
Front Immunol ; 12: 648754, 2021.
Article in English | MEDLINE | ID: mdl-33790913

ABSTRACT

Background and Aims: Emerging evidence has revealed that innate lymphoid cells (ILCs) play a key role in regulating metabolic disorders. Here, we investigated the role of group 3 ILCs (ILC3s) in the modulation of Non-alcoholic fatty liver disease (NAFLD). Methods: RORγ gfp/gfp (RORgt KI/KI) and Rag2-/- mice with the administration of A213, RORgt antagonist, fed with a high-fat-diet (HFD) for 12 weeks, were used. We performed flow cytometry, real time PCR, and lipidomics analysis of serum and liver, and used RAW264.7 cells and murine primary hepatocytes in vitro. Results: HFD increased ILC3s and M1 macrophages in the liver, and RORgt KI/KI mice deficient in ILC3 showed significant fatty liver, liver fibrosis and significantly increased palmitic acid levels in serum and liver. In addition, administration of A213 to Rag2-/- mice caused significant fatty liver, liver fibrosis, and a significant increase in serum and liver palmitate concentrations, as in RORgt KI/KI mice. Addition of palmitc acid stimulated IL-23 production in cell experiments using RAW264.7. IL-22 produced by ILC3s inhibited the palmitate-induced apoptosis of primary hepatocytes. Conclusions: HFD stimulates IL-23 production by M1 macrophages, thus promoting ILC3 proliferation, whereas IL-22 secreted by ILC3s contributes to the upregulation of hepatic lipid metabolism and has anti-apoptosis activity.


Subject(s)
Fatty Liver/immunology , Immunity, Innate/immunology , Liver/immunology , Lymphocytes/immunology , Macrophages/immunology , Animals , Apoptosis/immunology , Cells, Cultured , Diet, High-Fat/adverse effects , Fatty Liver/etiology , Fatty Liver/metabolism , Hepatocytes/cytology , Hepatocytes/immunology , Liver/metabolism , Liver/pathology , Lymphocytes/metabolism , Macrophages/classification , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Palmitic Acid/blood , Palmitic Acid/immunology , Palmitic Acid/metabolism , Protective Agents/metabolism , RAW 264.7 Cells
14.
Front Immunol ; 12: 641240, 2021.
Article in English | MEDLINE | ID: mdl-33833761

ABSTRACT

Metabolic Associated Fatty liver disease (MAFLD) is a global health problem and represents the most common cause of chronic liver disease in the world. MAFLD spectrum goes from simple steatosis to cirrhosis, in between metabolic steatohepatitis with progressive fibrosis, which pathogenesis is not completely understood. Hence, the role of the immune system has become an important fact in the trigger of inflammatory cascades in metabolic steatohepatitis and in the activation of hepatic stellate cells (HSCs). Among, the more studied immune cells in the pathogenesis of MAFLD are macrophages, T cells, natural killer and dendritic cells. In particular, hepatic dendritic cells had recently attracted a special attention, with a dual role in the pathogenesis of MAFLD. These cells have the capacity to switch from a tolerant state to active state inducing an inflammatory cascade. Furthermore, these cells play a role in the lipid storage within the liver, having, thus providing a crucial nexus between inflammation and lipid metabolism. In this review, we will discuss the current knowledge on the dual role of dendritic cells in lipid accumulation, as wells as in the triggering of hepatic inflammation and hepatocytes cell death in metabolic steatohepatitis.


Subject(s)
Dendritic Cells/immunology , Fatty Liver/immunology , Hepatic Stellate Cells/immunology , Lipid Metabolism/immunology , Liver/immunology , Macrophages/immunology , Animals , Dendritic Cells/pathology , Fatty Liver/pathology , Hepatic Stellate Cells/pathology , Humans , Inflammation/immunology , Inflammation/pathology , Liver/pathology , Macrophages/pathology
15.
Int J Mol Sci ; 22(4)2021 Feb 18.
Article in English | MEDLINE | ID: mdl-33670793

ABSTRACT

Liver transplantation (LTx) is often the only possible therapy for many end-stage liver diseases, but successful long-term transplant outcomes are limited by multiple factors, including ischemia reperfusion injury (IRI). This situation is aggravated by a shortage of transplantable organs, thus encouraging the use of inferior quality organs. Here, we have investigated early hepatic IRI in a retrospective, exploratory, monocentric case-control study considering organ marginality. We analyzed standard LTx biopsies from 46 patients taken at the end of cold organ preparation and two hours after reperfusion, and we showed that early IRI was present after two hours in 63% of cases. Looking at our data in general, in accordance with Eurotransplant criteria, a marginal transplant was allocated at our institution in about 54% of cases. We found that patients with a marginal-organ LTx showing evidence of IRI had a significantly worse one-year survival rate (51% vs. 75%). As we saw in our study cohort, the marginality of these livers was almost entirely due to steatosis. In contrast, survival rates in patients receiving a non-marginal transplant were not influenced by the presence or absence of IRI. Poorer outcomes in marginal organs prompted us to examine pre- and post-reperfusion biopsies, and it was revealed that transplants with IRI demonstrated significantly greater T cell infiltration. Molecular analyses showed that higher mRNA expression levels of CXCL-1, CD3 and TCRγ locus genes were found in IRI livers. We therefore conclude that the marginality of an organ, namely steatosis, exacerbates early IRI by enhancing effector immune cell infiltration. Preemptive strategies targeting immune pathways could increase the safety of using marginal organs for LTx.


Subject(s)
Fatty Liver/etiology , Fatty Liver/immunology , Liver Transplantation/adverse effects , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Reperfusion Injury/etiology , T-Lymphocytes/immunology , Allografts/pathology , Female , Gene Expression Regulation , Humans , Male , Middle Aged , Survival Analysis , Treatment Outcome
16.
Theranostics ; 11(7): 3489-3501, 2021.
Article in English | MEDLINE | ID: mdl-33537099

ABSTRACT

The rapid development and remarkable success of checkpoint inhibitors have provided significant breakthroughs in cancer treatment, including hepatocellular carcinoma (HCC). However, only 15-20% of HCC patients can benefit from checkpoint inhibitors. Cancer stem cells (CSCs) are responsible for recurrence, metastasis, and local and systemic therapy resistance in HCC. Accumulating evidence has suggested that HCC CSCs can create an immunosuppressive microenvironment through certain intrinsic and extrinsic mechanisms, resulting in immune evasion. Intrinsic evasion mechanisms mainly include activation of immune-related CSC signaling pathways, low-level expression of antigen presenting molecules, and high-level expression of immunosuppressive molecules. External evasion mechanisms are mainly related to HBV/HCV infection, alcoholic/nonalcoholic steatohepatitis, hypoxia stimulation, abnormal angiogenesis, and crosstalk between CSCs and immune cells. A better understanding of the complex mechanisms of CSCs involved in immune evasion will contribute to therapies for HCC. Here we will outline the detailed mechanisms of immune evasion for CSCs, and provide an overview of the current immunotherapies targeting CSCs in HCC.


Subject(s)
Carcinoma, Hepatocellular/therapy , Liver Neoplasms/therapy , Neoplasm Proteins/genetics , Neoplasm Recurrence, Local/therapy , Neoplastic Stem Cells/drug effects , Tumor Escape/drug effects , Antigen Presentation/drug effects , Antineoplastic Agents, Immunological/therapeutic use , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Fatty Liver/genetics , Fatty Liver/immunology , Fatty Liver/pathology , Fatty Liver/therapy , Gene Expression Regulation, Neoplastic , Hepatitis B/genetics , Hepatitis B/immunology , Hepatitis B/pathology , Hepatitis B/therapy , Hepatitis C/genetics , Hepatitis C/immunology , Hepatitis C/pathology , Hepatitis C/therapy , Humans , Immunologic Factors/therapeutic use , Immunotherapy/methods , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/immunology , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/prevention & control , Signal Transduction , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
17.
J Immunol ; 206(4): 904-916, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33441438

ABSTRACT

Age-related chronic inflammation promotes cellular senescence, chronic disease, cancer, and reduced lifespan. In this study, we wanted to explore the effects of a moderate exercise regimen on inflammatory liver disease and tumorigenesis. We used an established model of spontaneous inflammaging, steatosis, and cancer (nfkb1-/- mouse) to demonstrate whether 3 mo of moderate aerobic exercise was sufficient to suppress liver disease and cancer development. Interventional exercise when applied at a relatively late disease stage was effective at reducing tissue inflammation (liver, lung, and stomach), oxidative damage, and cellular senescence, and it reversed hepatic steatosis and prevented tumor development. Underlying these benefits were transcriptional changes in enzymes driving the conversion of tryptophan to NAD+, this leading to increased hepatic NAD+ and elevated activity of the NAD+-dependent deacetylase sirtuin. Increased SIRT activity was correlated with enhanced deacetylation of key transcriptional regulators of inflammation and metabolism, NF-κB (p65), and PGC-1α. We propose that moderate exercise can effectively reprogram pre-established inflammatory and metabolic pathologies in aging with the benefit of prevention of disease.


Subject(s)
Aging/immunology , Carcinogenesis/immunology , Fatty Liver/prevention & control , Liver Neoplasms/prevention & control , Physical Conditioning, Animal , Aging/genetics , Aging/pathology , Animals , Carcinogenesis/pathology , Cellular Senescence/immunology , Fatty Liver/immunology , Fatty Liver/pathology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Mice , Mice, Knockout , NF-kappa B p50 Subunit/genetics , NF-kappa B p50 Subunit/immunology
18.
Commun Biol ; 4(1): 15, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33398077

ABSTRACT

As a promising novel marine fish model for future research on marine ecotoxicology as well as an animal model of human disease, the genome information of yellowstripe goby (Mugilogobius chulae) remains unknown. Here we report the first annotated chromosome-level reference genome assembly for yellowstripe goby. A 20.67-cM sex determination region was discovered on chromosome 5 and seven potential sex-determining genes were identified. Based on combined genome and transcriptome data, we identified three key lipid metabolic pathways for high-fat accumulation in the liver of yellowstripe goby. The changes in the expression patterns of MGLL and CPT1 at different development stage of the liver, and the expansion of the ABCA1 gene, innate immune gene TLR23, and TRIM family genes may help in balancing high-fat storage in hepatocytes and steatohepatitis. These results may provide insights into understanding the molecular mechanisms of sex determination and high-fat storage in the liver of marine fishes.


Subject(s)
Lipogenesis , Liver/metabolism , Perciformes/genetics , Sex Determination Processes , ATP Binding Cassette Transporter 1 , Animals , Carnitine O-Palmitoyltransferase/metabolism , Fatty Liver/immunology , Female , Male , Monoacylglycerol Lipases/metabolism , Perciformes/immunology , Perciformes/metabolism , Phospholipids/biosynthesis , Whole Genome Sequencing
19.
Virulence ; 11(1): 1501-1511, 2020 12.
Article in English | MEDLINE | ID: mdl-33135589

ABSTRACT

Both hepatitis C virus (HCV) infection and retinol-binding protein 4 (RBP4) might contribute to insulin resistance (IR), how RBP4 links to IR in HCV infection remain elusive. A joint study of a prospective cohort of 842 chronically HCV-infected (CHC) patients (with 842 controls) and a line of HCV core transgenic mice was conducted. Of 842 patients, 771 had completed anti-HCV therapy and 667 had sustained virological responses (SVRs). Compared with controls, CHC patients had lower RBP4 levels. At baseline, age (95% CI ß: -0.87~-0.317), BMI (0.516~2.036), triglycerides (0.03~0.127), neutrophil-to-lymphocyte ratio (NLR) (1.561~7.327), and estimated glomerular filtration rate (eGFR) (-0.342~-0.149) levels were associated with RBP4 levels in CHC patients. At 24-week post-therapy, male sex (0.652~8.129), BMI (0.199~1.254), triglycerides (0.039~0.088), uric acid (0.599~3.067), eGFR (-0.247 ~-0.14) levels, and fibrosis-4 (-3.602~-0.039) scores were associated with RBP4 levels in SVR patients; compared with baseline, except genotype 3 HCV-infected patients, SVR patients had increased RBP4 levels, which were comparable with controls, while no HOMA-IR index alteration was noted after SVR. The HCV core transgenic mice exhibited nonobese hepatic steatosis, had higher hepatic RBP4 expression, higher serum levels of RBP4 and triglycerides, but comparable HOMA-IR levels than non-transgenic littermates. In conclusion, steatosis, sex, age, uric acid, NLR, and FIB-4 levels were associated with HCV-related RBP4 levels; BMI, triglycerides, and eGFR levels were associated with non-HCV-related RBP4 levels. Reversal of low RBP4 levels after SVR was evident in non-genotype 3 HCV-infected patients. Steatosis and inflammation linked with metabolic alteration other than IR, determined RBP4 levels in HCV-infected patients.


Subject(s)
Fatty Liver/virology , Hepacivirus/immunology , Hepatitis C, Chronic/immunology , Hepatitis C, Chronic/virology , Inflammation , Retinol-Binding Proteins, Plasma/genetics , Adult , Aged , Animals , Fatty Liver/immunology , Female , Hepacivirus/genetics , Humans , Male , Mice , Mice, Transgenic , Middle Aged , Prospective Studies , Retinol-Binding Proteins, Plasma/analysis , Retinol-Binding Proteins, Plasma/metabolism
20.
Arch Pharm Res ; 43(10): 997-1016, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33078304

ABSTRACT

Aberrant production of adipokines, a group of adipocytes-derived hormones, is considered one of the most important pathological characteristics of obesity. In individuals with obesity, beneficial adipokines, such as adiponectin are downregulated, whereas leptin and other pro-inflammatory adipokines are highly upregulated. Hence, the imbalance in levels of these adipokines is thought to promote the development of obesity-linked complications. However, the mechanisms by which adipokines contribute to the pathogenesis of various diseases have not been clearly understood. Inflammasomes represent key signaling platform that triggers the inflammatory and immune responses through the processing of the interleukin family of pro-inflammatory cytokines in a caspase-1-dependent manner. Beyond their traditional function as a component of the innate immune system, inflammasomes have been recently integrated into the pathological process of multiple metabolism- and obesity-related disorders such as cardiovascular diseases, diabetes, fatty liver disease, and cancer. Interestingly, emerging evidence also highlights the role of adipokines in the modulation of inflammasomes activation, making it a promising mechanism underlying distinct biological actions of adipokines in diseases driven by inflammation and metabolic disorders. In this review, we summarize the effects of adipokines, in particular adiponectin, leptin, visfatin and apelin, on inflammasomes activation and their implications in the pathophysiology of obesity-linked complications.


Subject(s)
Adipokines/metabolism , Inflammasomes/immunology , Metabolic Diseases/complications , Obesity/complications , Adipocytes/cytology , Adipocytes/immunology , Adipocytes/metabolism , Animals , Cardiovascular Diseases/immunology , Cardiovascular Diseases/metabolism , Diabetes Mellitus/immunology , Diabetes Mellitus/metabolism , Disease Models, Animal , Down-Regulation/immunology , Fatty Liver/immunology , Fatty Liver/metabolism , Humans , Inflammasomes/metabolism , Metabolic Diseases/immunology , Metabolic Diseases/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Obesity/immunology , Obesity/metabolism , Up-Regulation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...