Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
J Immunol ; 212(6): 974-981, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38251917

ABSTRACT

Activation and clonal expansion of the Ag-specific adaptive immune response in the draining lymph node is essential to clearing influenza A virus infections. Activation sufficient for virus clearance is dependent on the lymph node's architectural organization that is maintained by stromal cells, chiefly fibroblastic reticular cells. During an analysis of influenza A virus clearance in leptin receptor knockout (DB/DB) mice, we observed that the DB/DB mice have markedly reduced numbers of lymph node fibroblastic reticular cells at the steady state. The reduction in lymph node fibroblastic reticular cells resulted in abnormal lymph node organization and diminished numbers of adaptive immune cells in the lymph nodes under homeostatic conditions. As a consequence, the DB/DB mice were impaired in their ability to generate an effective influenza-specific adaptive immune response, which prevented virus clearance. Using leptin receptor mutant mice with point mutations at distinct signaling sites in the leptin receptor, we were able to link the leptin receptor's signaling domain tyrosine 985, which does not contribute to obesity, to lymph node fibroblastic reticular cell development and function. These results demonstrate a novel role for leptin receptor signaling in regulating lymph node development in a manner that is crucial to the generation of Ag-specific adaptive immune responses.


Subject(s)
Adaptive Immunity , Receptors, Leptin , Mice , Animals , Receptors, Leptin/genetics , Lymph Nodes , Signal Transduction , Mice, Inbred C57BL , Leptin
2.
J Virol ; 95(10)2021 04 26.
Article in English | MEDLINE | ID: mdl-33627392

ABSTRACT

Plasminogen activator inhibitor-1 (PAI-1) is a critical factor that regulates protein synthesis and degradation. The increased PAI-1 levels are detectable in the serum of patients with chronic hepatitis C virus (HCV) liver disease. The differentiation state and motility of HCV-induced cancer stem-like cells (CSC) play a major role in severe liver disease progression. However, the role of PAI-1 in the pathological process of chronic liver diseases remains unknown. In this study, we determined how PAI-1 affects the differentiation of CSC state in hepatocytes upon HCV infection. We found that HCV infection induced the expression of PAI-1 while decreasing miR-30c expression in Huh7.5.1 cells. Similar results were obtained from isolated hepatocytes from humanized liver mice after HCV infection. Moreover, decreased miR-30c expression in HCV-infected hepatocytes was associated with the increased levels of PAI-1 mRNA and protein. Notably, the increased PAI-1 levels resulted in the activation of Protein Kinase B/AKT, a major mediator of cell proliferation, in HCV-infected hepatocytes along with the increased expression of CSC markers such as Human Differentiated Protein (CD) 133, Epithelial cell adhesion molecule (EpCAM), Octamer 4 (Oct4), Nanog, Cyclin D1, and MYC. Moreover, blockade of PAI-1 activity by miR-30c mimic and anti-PAI-1 mAb abrogated the AKT activation with decreased expression of CSC markers. Our findings suggest that HCV infection induces the CSC state via PAI-1-mediated AKT activation in hepatocytes. It implicates that the manipulation of PAI-1 activity could provide potential therapeutics to prevent the development of HCV-associated chronic liver diseases.IMPORTANCEThe progression of chronic liver disease by HCV infection is considered a major risk factor for hepatocellular carcinoma (HCC), one of the major causes of death from cancer. Recent studies have demonstrated that increased CSC properties in HCV-infected hepatocytes are associated with the progression of HCC. Since proteins and miRNAs production by HCV-infected hepatocytes can play various roles in physiological processes, investigating these factors can potentially lead to new therapeutic targets. However, the mechanism of HCV associated progression of hepatocytes to CSC remains unclear. Here we identify the roles of PAI-1 and miR-30c in the progression of CSC during HCV infection in hepatocytes. Our data shows that increased secretion of PAI-1 following HCV infection promotes this CSC state and activation of AKT. We report that the inhibition of PAI-1 by miR-30c mimic reduces HCV associated CSC properties in hepatocytes. Taken together, targeting this interaction of secreted PAI-1 and miR-30c in HCV-infected hepatocytes may provide a potential therapeutic intervention against the progression to chronic liver diseases and HCC.

3.
Hepatology ; 74(6): 3409-3426, 2021 12.
Article in English | MEDLINE | ID: mdl-34218459

ABSTRACT

BACKGROUND AND AIMS: HCV infection is a major risk factor that can lead to chronic liver disease, including fibrosis, cirrhosis, and HCC. Progression of chronic liver disease by HCV infection is caused by a complex intercellular reaction. Especially, exosomes and microRNAs (miRNAs) from HCV-infected hepatocytes play a role in the pathogenesis of liver disease by facilitating cellular communication between parenchymal and nonparenchymal cells. However, the underlying mechanism of secretions of exosome and miRNAs during HCV infection is still open for study. APPROACH AND RESULTS: In this study, we demonstrated a pathway for the release of exosome and exosomal miRNAs through caspase-3/pannexin 1 (Panx1)/P2X4 activation during HCV infection in hepatocytes. We found that HCV infection induced the stimulation of exosome release and activation of the caspase-3/Panx1/P2X4 pathway in Huh7.5.1 cells. In addition, miR-122 and miR-146a levels in extracellular exosomes from HCV-infected cells were dramatically increased whereas intracellular miR122 and miR-146a expression had no large changes. Notably, secretions of exosomes and exosomal miRNAs were decreased by inhibition of caspase 3, Panx1, and P2X4 whereas inhibition of ROCK-1 cleavage did not affect these during HCV infection in Huh7.5.1 cells. CONCLUSIONS: These results suggested that HCV infection caused secretions of exosomes and exosomal miRNAs dependent on the caspase 3/Panx1/P2X4 pathway. Our study provides a possible therapeutic intervention using Panx1 suppression for liver disease development mediated by exosomes from HCV-infected hepatocytes.


Subject(s)
Connexins/metabolism , Hepatitis C, Chronic/genetics , Hepatocytes/virology , MicroRNAs/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Purinergic P2X4/metabolism , Blotting, Western , Cell Line , Exosomes/metabolism , Flow Cytometry , Hepatocytes/metabolism , Humans , Microscopy, Fluorescence , Real-Time Polymerase Chain Reaction
4.
J Biol Chem ; 294(40): 14591-14602, 2019 10 04.
Article in English | MEDLINE | ID: mdl-31375561

ABSTRACT

Zika virus (ZIKV)3 is an enveloped, single-stranded, positive-sense RNA virus of the Flaviviridae family that has emerged as a public health threat because of its global transmission and link to microcephaly. Currently there is no vaccine for this virus. Conversion of cholesterol to 25-hydroxycholesterol by cholesterol 25-hydroxylase (CH25H) has been shown to have broad antiviral properties. However, the molecular basis of induction of CH25H in humans is not known. Elucidation of signaling and transcriptional events for induction of CH25H expression is critical for designing therapeutic antiviral agents. In this study, we show that CH25H is induced by ZIKV infection or Toll-like receptor stimulation. Interestingly, CH25H is induced by pro-inflammatory cytokines, including IL-1ß, tumor necrosis factor α, and IL-6, and this induction depends on the STAT1 transcription factor. Additionally, we observed that cAMP-dependent transcription factor (ATF3) weakly binds to the CH25H promoter, suggesting cooperation with STAT1. However, ZIKV-induced CH25H was independent of type I interferon. These findings provide important information for understanding how the Zika virus induces innate inflammatory responses and promotes the expression of anti-viral CH25H protein.


Subject(s)
Activating Transcription Factor 3/genetics , STAT1 Transcription Factor/genetics , Steroid Hydroxylases/genetics , Zika Virus Infection/genetics , Zika Virus/genetics , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Cytokines/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Gene Expression Regulation, Enzymologic , Humans , Inflammation/enzymology , Inflammation/genetics , Inflammation/virology , Interferon Type I/genetics , Interleukin-1beta/genetics , Interleukin-6/genetics , Macrophages/virology , Steroid Hydroxylases/chemistry , Toll-Like Receptors/genetics , Tumor Necrosis Factor-alpha/genetics , Virus Replication/genetics , Zika Virus/pathogenicity , Zika Virus Infection/enzymology , Zika Virus Infection/virology
5.
Hepatology ; 67(1): 71-85, 2018 01.
Article in English | MEDLINE | ID: mdl-28749570

ABSTRACT

Hepatitis C virus (HCV) is a global health concern that can cause severe liver disease, such as cirrhosis and hepatocellular carcinoma. Control of HCV requires vigorous T-cell responses, yet CD4+ T cells in chronic HCV patients are dysfunctional. T follicular regulatory (Tfr) cells are a subset of regulatory T cells that suppress T follicular helper (Tfh) cells and the generation of high affinity antibody-producing B cells. In this study, we examined the accumulation of Tfr cells in the liver compartment during chronic HCV infection and defined the cellular and molecular mechanisms underlying their expansion. Our analysis revealed a substantial population of Tfr cells in livers of chronic HCV patients that is absent in liver tissues from nonviral hepatitis or healthy subjects. Coculture of PBMCs from healthy subjects with HCV-infected hepatoma cells resulted in preferential expansion of circulating Tfr cells, leading to suppression of Tfh cells. Additionally, coculture of tonsillar cells with infected hepatoma cells lead to an expansion of germinal center Tfr. Notably, expansion was mediated by transforming growth factor beta (TGF-ß)-containing exosomes released from HCV-infected hepatocytes given that blockade of exosome-associated TGF-ß or inhibition of exosome release abrogated Tfr expansion. CONCLUSION: These results show that liver-derived exosomes play a pivotal role in the accumulation of Tfr cells, likely leading to suppression of Tfh responses in HCV-infected patients. Our study identifies a novel pathway in which HCV infection in hepatocytes exacerbates Tfr cell responses to subvert antiviral immunity. (Hepatology 2018;67:71-85).


Subject(s)
Cell Proliferation/physiology , Exosomes/immunology , Hepatitis C/immunology , Hepatocytes/immunology , T-Lymphocytes, Regulatory/immunology , Biopsy, Needle , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Exosomes/metabolism , Flow Cytometry , Hepacivirus/immunology , Hepatitis C/pathology , Hepatocytes/metabolism , Humans , Immunohistochemistry , In Vitro Techniques , Statistics, Nonparametric , T-Lymphocytes, Regulatory/metabolism
6.
J Immunol ; 198(6): 2341-2351, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28159899

ABSTRACT

Cross-presentation is a modular series of intracellular events dictating the internalization and subsequent MHC class I (MHC I) display of extracellular Ags. This process has been defined in dendritic cells and plays a fundamental role in the induction of CD8+ T cell immunity during viral, intracellular bacterial, and antitumor responses. Herein, acute viral infection of murine liver with adenovirus, a model for intrahepatic cross-presentation, confirms hepatocytes directly contribute to cross-presentation of Ags and priming the pool of naive CD8+ T cells within the liver microenvironment. Processing of soluble and cell-associated Ags into peptide displayed by MHC I is however defective in hepatocytes lacking collectrin, an intracellular chaperone protein that localizes within the endoplasmic reticulum-Golgi intermediate compartment. Loss of hepatic collectrin expression leads to the diminished cross-priming and expansion of cytolytic antiviral CD8+ T cells. This study demonstrates that collectrin positively regulates processing of engulfed Ags into MHC I:peptide complexes within hepatocytes. Collectrin-mediated cross-presentation supports intrahepatic adaptive antiviral immune responses and may lead to insights into the nature of how the liver acts as a primary site of CD8+ T cell activation.


Subject(s)
Adenoviridae Infections/immunology , Adenoviridae/immunology , CD8-Positive T-Lymphocytes/immunology , Cross-Priming , Hepatocytes/immunology , Liver/immunology , Membrane Glycoproteins/metabolism , Acute Disease , Animals , Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/virology , Extracellular Space/immunology , Hepatocytes/virology , Histocompatibility Antigens Class I/metabolism , Liver/virology , Lymphocyte Activation/genetics , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Solubility , Transplantation Chimera
7.
J Immunol ; 198(7): 2589-2601, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28219886

ABSTRACT

Glomerular damage mediated by glomerulus-infiltrating myeloid-derived cells is a key pathogenic event in lupus nephritis (LN), but the process is poorly understood. Confocal microscopy of kidney sections and flow cytometry analysis of glomerular cells from magnetic bead-purified glomeruli have identified glomerulus-infiltrating leukocyte populations in NZM2328 (NZM) lupus-prone mice with spontaneous chronic glomerulonephritis (GN) and anti-glomerular basement membrane-induced nephritis. The occurrence of a major glomerulus-infiltrating CD11b+F4/80-I-A- macrophage population exhibiting the markers programmed death ligand-1 (PD-L1), Mac-2, and macrophage mannose receptor (CD206) and producing Klf4, Il10, Retnla, Tnf, and Il6 mRNA, which are known to be expressed by alternatively activated (M2b) macrophages, correlated with proteinuria status. In NZM mice with spontaneous LN, glomerular macrophage infiltration is predominant. CD11b+F4/80-I-A- intraglomerular macrophages and polymorphonuclear neutrophils (PMN) are important in inducing GN, as anti-CD11b and -ICAM-1 mAb inhibited both proteinuria and macrophage and PMN infiltration. The predominant and high expression of PD-L1 by CD11b+F4/80-I-A- glomerular macrophages in kidneys of mice with GN and the inhibition of proteinuria by anti-PD-L1 mAb supported the pathogenic role of these macrophages but not the PD-L1- PMN in GN development and in inducing podocyte damage. In NZM mice with spontaneous chronic GN and severe proteinuria, few glomerulus-infiltrating PMN were found, leaving macrophages and, to a less extent, dendritic cells as the major infiltrating leukocytes. Taken together, these data support the important pathogenic effect of CD11b+F4/80-I-A- M2b-like glomerulus-infiltrating macrophages in LN and reinforce macrophages as a promising target for GN treatment.


Subject(s)
Kidney Glomerulus/immunology , Lupus Nephritis/immunology , Macrophages/immunology , Animals , B7-H1 Antigen/immunology , Bone Marrow Cells/immunology , Cell Separation , Disease Models, Animal , Female , Flow Cytometry , Kidney Glomerulus/pathology , Kruppel-Like Factor 4 , Lupus Nephritis/pathology , Macrophage-1 Antigen/immunology , Macrophages/pathology , Mice , Mice, Mutant Strains , Microscopy, Confocal , Real-Time Polymerase Chain Reaction
8.
Hepatology ; 65(1): 32-43, 2017 01.
Article in English | MEDLINE | ID: mdl-27770558

ABSTRACT

The liver maintains an immunologically tolerant environment as a result of continuous exposure to food and bacterial constituents from the digestive tract. Hepatotropic pathogens can take advantage of this niche and establish lifelong chronic infections causing hepatic fibrosis and hepatocellular carcinoma. Macrophages (Mϕ) play a critical role in regulation of immune responses to hepatic infection and regeneration of tissue. However, the factors crucial for Mϕ in limiting hepatic inflammation or resolving liver damage have not been fully understood. In this report, we demonstrate that expression of C-type lectin receptor scavenger receptor-AI (SR-AI) is crucial for promoting M2-like Mϕ activation and polarization during hepatic inflammation. Liver Mϕ uniquely up-regulated SR-AI during hepatotropic viral infection and displayed increased expression of alternative Mϕ activation markers, such as YM-1, arginase-1, and interleukin-10 by activation of mer receptor tyrosine kinase associated with inhibition of mammalian target of rapamycin. Expression of these molecules was reduced on Mϕ obtained from livers of infected mice deficient for the gene encoding SR-AI (msr1). Furthermore, in vitro studies using an SR-AI-deficient Mϕ cell line revealed impeded M2 polarization and decreased phagocytic capacity. Direct stimulation with virus was sufficient to activate M2 gene expression in the wild-type (WT) cell line, but not in the knockdown cell line. Importantly, tissue damage and fibrosis were exacerbated in SR-AI-/- mice following hepatic infection and adoptive transfer of WT bone-marrow-derived Mϕ conferred protection against fibrosis in these mice. CONCLUSION: SR-AI expression on liver Mϕ promotes recovery from infection-induced tissue damage by mediating a switch to a proresolving Mϕ polarization state. (Hepatology 2017;65:32-43).


Subject(s)
Hepatitis/etiology , Liver Cirrhosis/etiology , Macrophage Activation , Scavenger Receptors, Class A/biosynthesis , Animals , Cells, Cultured , Female , Mice , Mice, Inbred C57BL
9.
J Immunol ; 196(5): 2283-92, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26826241

ABSTRACT

The hepatitis C virus (HCV) infects ∼ 200 million people worldwide. The majority of infected individuals develop persistent infection, resulting in chronic inflammation and liver disease, including cirrhosis and hepatocellular carcinoma. The ability of HCV to establish persistent infection is partly due to its ability to evade the immune response through multiple mechanisms, including suppression of NK cells. NK cells control HCV replication during the early phase of infection and regulate the progression to chronic disease. In particular, IFN-γ produced by NK cells limits viral replication in hepatocytes and is important for the initiation of adaptive immune responses. However, NK cell function is significantly impaired in chronic HCV patients. The cellular and molecular mechanisms responsible for impaired NK cell function in HCV infection are not well defined. In this study, we analyzed the interaction of human NK cells with CD33(+) PBMCs that were exposed to HCV. We found that NK cells cocultured with HCV-conditioned CD33(+) PBMCs produced lower amounts of IFN-γ, with no effect on granzyme B production or cell viability. Importantly, this suppression of NK cell-derived IFN-γ production was mediated by CD33(+)CD11b(lo)HLA-DR(lo) myeloid-derived suppressor cells (MDSCs) via an arginase-1-dependent inhibition of mammalian target of rapamycin activation. Suppression of IFN-γ production was reversed by l-arginine supplementation, consistent with increased MDSC arginase-1 activity. These novel results identify the induction of MDSCs in HCV infection as a potent immune evasion strategy that suppresses antiviral NK cell responses, further indicating that blockade of MDSCs may be a potential therapeutic approach to ameliorate chronic viral infections in the liver.


Subject(s)
Arginase/metabolism , Hepacivirus/immunology , Interferon-gamma/biosynthesis , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Myeloid Cells/immunology , Myeloid Cells/metabolism , Arginine/metabolism , Cell Line , Cells, Cultured , Hepatitis C/genetics , Hepatitis C/immunology , Hepatitis C/metabolism , Hepatitis C/virology , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , Myeloid Cells/virology , RNA Processing, Post-Transcriptional , Sialic Acid Binding Ig-like Lectin 3/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
10.
Hepatology ; 63(3): 799-812, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26662852

ABSTRACT

UNLABELLED: Nonalcoholic steatohepatitis (NASH) affects 3%-5% of the U.S. population, having severe clinical complications to the development of fibrosis and end-stage liver diseases, such as cirrhosis and hepatocellular carcinoma. A critical cause of NASH is chronic systemic inflammation promoted by innate immune cells, such as liver macrophages (Mϕ) and natural killer (NK) cells. However, little is known about how the crosstalk between Mϕ and NK cells contributes to regulate NASH progression to fibrosis. In this report, we demonstrate that NKp46(+) cells play an important role in preventing NASH progression to fibrosis by regulating M1/M2 polarization of liver Mϕ. Using a murine model of NASH, we demonstrate that DX5(+)NKp46(+) NK cells are increased during disease and play a role in polarizing Mϕ toward M1-like phenotypes. This NK's immunoregulatory function depends on the production of interferon-gamma (IFN-γ), but not by granzyme-mediated cytolytic activity. Notably, depletion of NKp46(+) cells promotes the development of fibrosis with increased expression of profibrogenic genes as well as skewed M2 Mϕ phenotypes in hepatic tissues. CONCLUSIONS: NK cell-derived IFN-γ may be essential for maintaining a balanced inflammatory environment that promotes tissue integrity and limiting NASH progression to fibrosis.


Subject(s)
Killer Cells, Natural/physiology , Liver/immunology , Macrophages/physiology , Non-alcoholic Fatty Liver Disease/immunology , Transforming Growth Factor beta/metabolism , Animals , Cell Death , Collagen/metabolism , Disease Progression , Female , Fibrosis , Hepatic Stellate Cells/metabolism , Immunity, Innate , Interferon-gamma/metabolism , Liver/pathology , Macrophage Activation , Mice, Inbred C57BL , Natural Cytotoxicity Triggering Receptor 1/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Phenotype , Receptor Cross-Talk , T-Lymphocytes, Regulatory
11.
Crit Rev Immunol ; 36(5): 379-394, 2016.
Article in English | MEDLINE | ID: mdl-28605345

ABSTRACT

Understanding of antigen-presenting cell (APC) participation in tissue inflammation and metabolism has advanced through numerous studies using systems biology approaches. Previously unrecognized connections between these research areas have been elucidated in the context of inflammatory disease involving innate and adaptive immune responses. A new conceptual framework bridges APC biology, metabolism, and cytokines in the generation of effective T-cell responses. Exploring these connections is paramount to addressing the rising tide of multi-organ system diseases, particularly chronic diseases associated with metabolic syndrome, infection, and cancer. Focused research in these areas will aid the development of strategies to harness and manipulate innate immunology to improve vaccine development, anti-viral, anti-inflammatory, and anti-tumor therapies. This review highlights recent advances in APC "immunometabolism" specifically related to chronic viral and metabolic disease in humans. The goal of this review is to develop an abridged and consolidated outlook on recent thematic updates to APC immunometabolism in the areas of regulation and crosstalk between metabolic and inflammatory signaling and the integrated stress response and how these signals dictate APC function in providing T-cell activation Signal 3.


Subject(s)
Dendritic Cells/immunology , Macrophages/immunology , T-Lymphocytes/immunology , Animals , Humans , Interferons , Signal Transduction
12.
J Immunol ; 194(7): 3213-22, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25712214

ABSTRACT

The liver maintains a tolerogenic environment to avoid unwarranted activation of its resident immune cells upon continuous exposure to food and bacterially derived Ags. However, in response to hepatotropic viral infection, the liver's ability to switch from a hyporesponsive to a proinflammatory environment is mediated by select sentinels within the parenchyma. To determine the contribution of hepatic dendritic cells (DCs) in the activation of naive CD8(+) T cells, we first characterized resident DC subsets in the murine liver. Liver DCs exhibit unique properties, including the expression of CD8α (traditionally lymphoid tissue specific), CD11b, and CD103 markers. In both the steady-state and following viral infection, liver CD103(+) DCs express high levels of MHC class II, CD80, and CD86 and contribute to the high number of activated CD8(+) T cells. Importantly, viral infection in the Batf3(-/-) mouse, which lacks CD8α(+) and CD103(+) DCs in the liver, results in a 3-fold reduction in the proliferative response of Ag-specific CD8(+) T cells. Limiting DC migration out of the liver does not significantly alter CD8(+) T cell responsiveness, indicating that CD103(+) DCs initiate the induction of CD8(+) T cell responses in situ. Collectively, these data suggest that liver-resident CD103(+) DCs are highly immunogenic in response to hepatotropic viral infection and serve as a major APC to support the local CD8(+) T cell response. It also implies that CD103(+) DCs present a promising cellular target for vaccination strategies to resolve chronic liver infections.


Subject(s)
Antigens, CD/metabolism , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Integrin alpha Chains/metabolism , Liver/immunology , Lymphocyte Activation/immunology , Adenoviridae/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigens, Surface/metabolism , CD11b Antigen/metabolism , Cell Movement , Female , Immunophenotyping , Liver/pathology , Liver/virology , Male , Mice , Mice, Transgenic , Phenotype , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Viruses/immunology
13.
Immunol Rev ; 255(1): 210-21, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23947357

ABSTRACT

Myeloid derived suppressor cells (MDSCs) are immature cells of myeloid origin, frequently found in tumor microenvironments and in the blood of cancer patients. In recent years, MDSCs have also been found in non-cancer settings, including a number of viral infections. The evasion of host immunity employed by viruses to establish viral persistence strikingly parallels mechanisms of tumor escape, prompting investigations into the generation and function of MDSCs in chronic viral infections. Importantly, analogous to the tumor microenvironment, MDSCs effectively suppress antiviral host immunity by limiting the function of several immune cells including T cells, natural killer cells, and antigen-presenting cells. In this article, we review studies on the mechanisms of MDSC generation, accumulation, and survival in an effort to understand their emergent importance in viral infections. We include a growing list of viral infections in which MDSCs have been reported. Finally, we discuss how MDSCs might play a role in establishing chronic viral infections and identify potential therapeutics that target MDSCs.


Subject(s)
Myeloid Cells/immunology , Virus Diseases/immunology , Viruses/immunology , Animals , Epigenesis, Genetic , Humans , Immune Tolerance , Immunotherapy , Myeloid Cells/cytology , Myeloid Cells/metabolism , Virus Diseases/genetics , Virus Diseases/metabolism , Virus Diseases/therapy
14.
J Biol Chem ; 289(8): 5310-9, 2014 Feb 21.
Article in English | MEDLINE | ID: mdl-24385435

ABSTRACT

Hepatitis C virus (HCV) infection in hepatocytes stimulates innate antiviral responses including the production of type III interferons (IFN-λ), including IL-28A, IL-28B, and IL-29. However, the molecular mechanism(s) regulating the expression of IFN-λ genes in HCV-infected hepatocytes remains undefined. In this study, we examined regulatory elements involved in the induction of IFN-λ genes following HCV infection in hepatocytes and further determined the binding of specific transcription factor(s) to promoter regions of IFN-λ genes. Our studies reveal that the regulatory portion for IL-28A, IL-28B, and IL-29 genes is localized to a 1-kb region in their respective promoters. Notably, interferon regulatory factor (IRF)-3 and -7 are the key transcriptional factors for the induction of IL-28A and IL-28B genes, whereas NF-κB is an additional requirement for the induction of the IL-29 gene. Ligation of Toll-like receptors (TLR) 3, 7, 8, and 9, which also activate IRFs and NF-κB, resulted in more robust production of IFN-λ than that observed with HCV infection, verifying the importance of TLR pathways in IFN-λ production. Furthermore, the addition of IFN-λ to HCV-infected hepatocytes decreased viral replication and produced a concurrent reduction in microRNA-122 (miR-122). The decrease in viral replication was enhanced by the co-administration of IFN-λ and miR-122 inhibitor (miRIDIAN), suggesting that such combinatorial therapies may be beneficial for the treatment of chronic HCV infection.


Subject(s)
Gene Expression Regulation , Hepacivirus/physiology , Hepatocytes/virology , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-7/metabolism , Interleukins/genetics , NF-kappa B/metabolism , Cells, Cultured , Hepatitis C/genetics , Hepatitis C/virology , Hepatocytes/metabolism , Humans , Interferons , Interleukins/metabolism , Ligands , Liver/metabolism , Liver/pathology , Liver/virology , MicroRNAs/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , Transcription, Genetic , Transcriptional Activation/genetics
15.
PLoS Pathog ; 9(9): e1003615, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24068930

ABSTRACT

Respiratory virus infections, such as influenza, typically induce a robust type I (pro-inflammatory cytokine) immune response, however, the production of type 2 cytokines has been observed. Type 2 cytokine production during respiratory virus infection is linked to asthma exacerbation; however, type 2 cytokines may also be tissue protective. Interleukin (IL)-5 is a prototypical type 2 cytokine that is essential for eosinophil maturation and egress out of the bone marrow. However, little is known about the cellular source and underlying cellular and molecular basis for the regulation of IL-5 production during respiratory virus infection. Using a mouse model of influenza virus infection, we found a robust transient release of IL-5 into infected airways along with a significant and progressive accumulation of eosinophils into the lungs, particularly during the recovery phase of infection, i.e. following virus clearance. The cellular source of the IL-5 was group 2 innate lymphoid cells (ILC2) infiltrating the infected lungs. Interestingly, the progressive accumulation of eosinophils following virus clearance is reflected in the rapid expansion of c-kit⁺ IL-5 producing ILC2. We further demonstrate that the enhanced capacity for IL-5 production by ILC2 during recovery is concomitant with the enhanced expression of the IL-33 receptor subunit, ST2, by ILC2. Lastly, we show that NKT cells, as well as alveolar macrophages (AM), are endogenous sources of IL-33 that enhance IL-5 production from ILC2. Collectively, these results reveal that c-kit⁺ ILC2 interaction with IL-33 producing NKT and AM leads to abundant production of IL-5 by ILC2 and accounts for the accumulation of eosinophils observed during the recovery phase of influenza infection.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Interleukin-5/metabolism , Killer Cells, Natural/immunology , Lung/immunology , Macrophages, Alveolar/immunology , Orthomyxoviridae Infections/immunology , Respiratory Tract Infections/immunology , Animals , Cells, Cultured , Eosinophilia/etiology , Eosinophils/immunology , Eosinophils/metabolism , Eosinophils/pathology , Eosinophils/virology , Immunity, Innate , Interleukin-33 , Interleukins/metabolism , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Killer Cells, Natural/virology , Lung/metabolism , Lung/pathology , Lung/virology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Macrophages, Alveolar/virology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Protein Subunits/metabolism , Receptors, Interleukin/metabolism , Respiratory Tract Infections/metabolism , Respiratory Tract Infections/pathology , Respiratory Tract Infections/virology , Specific Pathogen-Free Organisms , Up-Regulation
16.
Hepatology ; 59(4): 1351-65, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24677194

ABSTRACT

UNLABELLED: The liver is a tolerogenic environment exploited by persistent infections, such as hepatitis B (HBV) and C (HCV) viruses. In a murine model of intravenous hepatotropic adenovirus infection, liver-primed antiviral CD8(+) T cells fail to produce proinflammatory cytokines and do not display cytolytic activity characteristic of effector CD8(+) T cells generated by infection at an extrahepatic, that is, subcutaneous, site. Importantly, liver-generated CD8(+) T cells also appear to have a T-regulatory (Treg) cell function exemplified by their ability to limit proliferation of antigen-specific T-effector (Teff ) cells in vitro and in vivo via T-cell immunoglobulin and mucin 3 (Tim-3) expressed by the CD8(+) Treg cells. Regulatory activity did not require recognition of the canonical Tim-3 ligand, galectin-9, but was dependent on CD8(+) Treg cell-surface Tim-3 binding to the alarmin, high-mobility group box 1 (HMGB-1). CONCLUSION: Virus-specific Tim-3(+) CD8(+) T cells operating through HMGB-1 recognition in the setting of acute and chronic viral infections of the liver may act to dampen hepatic T-cell responses in the liver microenvironment and, as a consequence, limit immune-mediated tissue injury or promote the establishment of persistent infections.


Subject(s)
Adaptive Immunity/physiology , Adenoviridae Infections/immunology , Adenoviridae Infections/physiopathology , CD8-Positive T-Lymphocytes/physiology , Galectins/physiology , HMGB1 Protein/physiology , Mucin-3/physiology , Adenoviridae/physiology , Adenoviridae Infections/pathology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Proliferation , Cellular Microenvironment , Disease Models, Animal , Immunoglobulins/physiology , In Vitro Techniques , Liver/pathology , Liver/physiopathology , Liver/virology , Mice , Mice, Inbred Strains , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
18.
Hepatology ; 57(4): 1314-24, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23150092

ABSTRACT

UNLABELLED: Hepatitis C virus (HCV) is a major cause of liver cirrhosis and hepatocellular carcinoma. Here we report that infection of hepatic cells by HCV stimulates nuclear factor kappa B (NFκB)-dependent production of thymic stromal lymphopoietin (TSLP). Hepatocyte-derived TSLP in turn conditions dendritic cells (DCs) to drive T-helper (Th)17 differentiation. The TSLP secreted by HCV-infected hepatoma cells is capable of activating human monocyte-derived DCs by up-regulating the expression of CD40, CD86, CCL17, CCL22, and CCL20 which are activating markers of DCs. In addition, the production of key cytokines for Th17 differentiation, transforming growth factor beta (TGF-ß), interleukin (IL)-6, and IL-21, is enhanced by human monocytes upon coculture with HCV-infected cells. Importantly, the blockade of TSLP using neutralizing antibody prevented the activation and maturation of DCs as well as the production of Th17 differentiation cytokines. DC conditioning by TSLP secreted from HCV-infected cells activated naïve CD4+ T lymphocytes, resulting in Th17 differentiation. Furthermore, we can detect substantial levels of hepatocyte TSLP in fibrotic liver tissue from chronic HCV patients. Thus, blockade of TSLP released by HCV-infected hepatocytes may suppress the induction/maintenance of hepatic Th17 responses and halt the progression of chronic liver disease to fibrosis and liver failure. CONCLUSION: Hepatocyte-derived TSLP conditions DCs to drive Th17 differentiation. Treatment of TSLP neutralizing antibody in HCV-infected hepatocyte/DC coculture abrogates DC conditioning and thereby inhibits Th17 differentiation.


Subject(s)
CD4-Positive T-Lymphocytes/pathology , Cell Differentiation , Cytokines/metabolism , Hepacivirus/physiology , Hepatocytes/metabolism , Hepatocytes/virology , Th17 Cells/pathology , Antibodies, Neutralizing/pharmacology , CD4-Positive T-Lymphocytes/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Cell Communication , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Cytokines/antagonists & inhibitors , Cytokines/drug effects , Dendritic Cells/pathology , Hepatitis C/pathology , Hepatocytes/pathology , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/virology , Monocytes/pathology , NF-kappa B/metabolism , Th17 Cells/metabolism , Thymic Stromal Lymphopoietin
19.
PLoS One ; 19(6): e0306345, 2024.
Article in English | MEDLINE | ID: mdl-38935609

ABSTRACT

Chronic liver diseases are caused by hepatic viral infection, chemicals, and metabolic stress. The protein Grb2-associated binder 1 (Gab1) binds to various growth factor receptors, and triggers cell differentiation/survival signaling pathways. To identify signaling molecules involved in the progression of liver diseases, we performed reverse-phase protein microarray (RPMA)-based screening of hepatocytes isolated from humanized mice after acute HCV infection. Acute viral infection in humanized liver mice significantly decreased the level of hepatocyte p-Gab1. Moreover, hepatoma cells upon HCV infection decreased Gab1 mRNA at later times of infection (D3 to D5) and p-Gab1 level was inversely related to the production of TGF-ß. In contrast, the level of p-Gab1 was increased in CCL4-induced fibrotic liver. Hepatoma cells showed elevation of p-Gab1, along with an increase in STAT3 and ERK activation, upon treatment with HGF (ligand of HGF receptor/c-Met) and CCL4. In Gab1 knockdown hepatoma cells, cell proliferative signaling activity was reduced but the level of activated caspase-3 was increased. These findings suggest that hepatocyte Gab1 expression may play a role in promoting liver fibrosis progression by triggering ERK activation and inhibiting apoptosis. It implies that the Gab1-mediated signaling pathway would be a promising therapeutic target to treat chronic liver diseases.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis , Cell Proliferation , Hepatocyte Growth Factor , Hepatocytes , Liver Cirrhosis , Proto-Oncogene Proteins c-met , Signal Transduction , Animals , Hepatocytes/metabolism , Hepatocytes/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Humans , Mice , Proto-Oncogene Proteins c-met/metabolism , Hepatocyte Growth Factor/metabolism , Cell Line, Tumor , Hepatitis C/metabolism , Hepatitis C/pathology , Hepatitis C/complications
20.
J Biol Chem ; 287(48): 40161-72, 2012 Nov 23.
Article in English | MEDLINE | ID: mdl-23066023

ABSTRACT

BACKGROUND: The mechanisms triggering nonalcoholic steatohepatitis (NASH) remain poorly defined. RESULTS: Kupffer cells are the first responding cells to hepatocyte injuries, leading to TNFα production, chemokine induction, and monocyte recruitment. The silencing of TNFα in myeloid cells reduces NASH progression. CONCLUSION: Increase of TNFα-producing Kupffer cells is crucial for triggering NASH via monocyte recruitment. SIGNIFICANCE: Myeloid cells-targeted silencing of TNFα might be a tenable therapeutic approach. Nonalcoholic steatohepatitis (NASH), characterized by lipid deposits within hepatocytes (steatosis), is associated with hepatic injury and inflammation and leads to the development of fibrosis, cirrhosis, and hepatocarcinoma. However, the pathogenic mechanism of NASH is not well understood. To determine the role of distinct innate myeloid subsets in the development of NASH, we examined the contribution of liver resident macrophages (i.e. Kupffer cells) and blood-derived monocytes in triggering liver inflammation and hepatic damage. Employing a murine model of NASH, we discovered a previously unappreciated role for TNFα and Kupffer cells in the initiation and progression of NASH. Sequential depletion of Kupffer cells reduced the incidence of liver injury, steatosis, and proinflammatory monocyte infiltration. Furthermore, our data show a differential contribution of Kupffer cells and blood monocytes during the development of NASH; Kupffer cells increased their production of TNFα, followed by infiltration of CD11b(int)Ly6C(hi) monocytes, 2 and 10 days, respectively, after starting the methionine/choline-deficient (MCD) diet. Importantly, targeted knockdown of TNFα expression in myeloid cells decreased the incidence of NASH development by decreasing steatosis, liver damage, monocyte infiltration, and the production of inflammatory chemokines. Our findings suggest that the increase of TNFα-producing Kupffer cells in the liver is crucial for the early phase of NASH development by promoting blood monocyte infiltration through the production of IP-10 and MCP-1.


Subject(s)
Fatty Liver/immunology , Kupffer Cells/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Choline/metabolism , Diet/adverse effects , Disease Models, Animal , Fatty Liver/etiology , Fatty Liver/genetics , Fatty Liver/metabolism , Female , Humans , Methionine/deficiency , Mice , Mice, Inbred C57BL , Monocytes/immunology , Non-alcoholic Fatty Liver Disease , Tumor Necrosis Factor-alpha/genetics
SELECTION OF CITATIONS
SEARCH DETAIL