Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
Add more filters

Country/Region as subject
Publication year range
1.
PLoS Pathog ; 17(8): e1009799, 2021 08.
Article in English | MEDLINE | ID: mdl-34370798

ABSTRACT

Chronic infection with HCV is manifested by dysregulation of innate immune responses and impaired T cell function at multiple levels. These changes may impact susceptibility to other infections, responsiveness to antiviral therapies, vaccine responsiveness, and development of complications such as hepatocellular carcinoma. Highly effective direct-acting antiviral (DAA) therapy has revolutionized the management of chronic HCV, with expected cure rates exceeding 95%. DAA treatment represents a unique opportunity to investigate to what extent elimination of viral replication and chronic antigen stimulation can restore immunologic phenotype. In this study we interrogated the global transcriptional profile of isolated peripheral blood T cells before, during and after IFN-free DAA therapy using single-cell mRNA sequencing. Our results demonstrate that T cells mapped at single-cell resolution have dramatic transcriptomic changes early after initiation of DAA and many of these changes are sustained after completion of DAA therapy. Specifically, we see a significant reduction in transcripts associated with innate immune activation and interferon signaling such as ISG15, ISG20, IFIT3, OAS and MX1 in many different T cell subsets. Furthermore, we find an early upregulation of a gene involved in suppression of immune activation, DUSP1, in circulating T cells. Conclusion: This study provides the first in-depth transcriptomic analysis at the single-cell level of patients undergoing DAA therapy, demonstrating that IFN-free antiviral therapy in chronic HCV infection induces hitherto unrecognized shifts in innate immune and interferon signaling within T cell populations early, during, and long-term after treatment. The present study provides a rich data source to explore the effects of DAA treatment on bulk T cells.


Subject(s)
Antiviral Agents/therapeutic use , Gene Expression Regulation/drug effects , Hepatitis C, Chronic/genetics , Interferons/genetics , Single-Cell Analysis/methods , T-Lymphocyte Subsets/metabolism , Transcriptome/drug effects , Biomarkers/blood , Hepacivirus/drug effects , Hepacivirus/physiology , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/virology , Humans , Interferons/metabolism , Male , Prospective Studies , T-Lymphocyte Subsets/drug effects
2.
Immunity ; 40(1): 66-77, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24412613

ABSTRACT

Acute intestinal inflammation involves early accumulation of neutrophils (PMNs) followed by either resolution or progression to chronic inflammation. Based on recent evidence that mucosal metabolism influences disease outcomes, we hypothesized that transmigrating PMNs influence the transcriptional profile of the surrounding mucosa. Microarray studies revealed a cohort of hypoxia-responsive genes regulated by PMN-epithelial crosstalk. Transmigrating PMNs rapidly depleted microenvironmental O2 sufficiently to stabilize intestinal epithelial cell hypoxia-inducible factor (HIF). By utilizing HIF reporter mice in an acute colitis model, we investigated the relative contribution of PMNs and the respiratory burst to "inflammatory hypoxia" in vivo. CGD mice, lacking a respiratory burst, developed accentuated colitis compared to control, with exaggerated PMN infiltration and diminished inflammatory hypoxia. Finally, pharmacological HIF stabilization within the mucosa protected CGD mice from severe colitis. In conclusion, transcriptional imprinting by infiltrating neutrophils modulates the host response to inflammation, via localized O2 depletion, resulting in microenvironmental hypoxia and effective inflammatory resolution.


Subject(s)
Colitis/immunology , Hypoxia/immunology , Mucous Membrane/metabolism , Neutrophils/pathology , Animals , Cell Communication , Cell Movement , Cells, Cultured , Cellular Microenvironment , Colitis/chemically induced , Colon/pathology , Disease Models, Animal , Hypoxia/chemically induced , Hypoxia-Inducible Factor 1/metabolism , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , Mucous Membrane/pathology , NADPH Oxidase 2 , NADPH Oxidases/genetics , Oxidative Stress , Oxygen/metabolism , Protein Stability/drug effects , Transendothelial and Transepithelial Migration
3.
Hepatology ; 74(3): 1546-1559, 2021 09.
Article in English | MEDLINE | ID: mdl-33730435

ABSTRACT

BACKGROUND AND AIMS: Nonalcoholic fatty liver disease (NAFLD) is the most prevalent cause of liver disease in children. Mercury (Hg), a ubiquitous toxic metal, has been proposed as an environmental factor contributing to toxicant-associated fatty liver disease. APPROACH AND RESULTS: We investigated the effect of prenatal exposure to Hg on childhood liver injury by combining epidemiological results from a multicenter mother-child cohort with complementary in vitro experiments on monocyte cells that are known to play a key role in liver immune homeostasis and NAFLD. We used data from 872 mothers and their children (median age, 8.1 years; interquartile range [IQR], 6.5-8.7) from the European Human Early-Life Exposome cohort. We measured Hg concentration in maternal blood during pregnancy (median, 2.0 µg/L; IQR, 1.1-3.6). We also assessed serum levels of alanine aminotransferase (ALT), a common screening tool for pediatric NAFLD, and plasma concentrations of inflammation-related cytokines in children. We found that prenatal Hg exposure was associated with a phenotype in children that was characterized by elevated ALT (≥22.1 U/L for females and ≥25.8 U/L for males) and increased concentrations of circulating IL-1ß, IL-6, IL-8, and TNF-α. Consistently, inflammatory monocytes exposed in vitro to a physiologically relevant dose of Hg demonstrated significant up-regulation of genes encoding these four cytokines and increased concentrations of IL-8 and TNF-α in the supernatants. CONCLUSIONS: These findings suggest that developmental exposure to Hg can contribute to inflammation and increased NAFLD risk in early life.


Subject(s)
Mercury/blood , Non-alcoholic Fatty Liver Disease/epidemiology , Prenatal Exposure Delayed Effects/epidemiology , Adult , Alanine Transaminase , Child , Cohort Studies , Cytokines , Disease Susceptibility , Exposome , Female , Humans , Inflammation , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Male , Maternal Exposure , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Prenatal Exposure Delayed Effects/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
4.
Hepatology ; 70(1): 67-83, 2019 07.
Article in English | MEDLINE | ID: mdl-30516830

ABSTRACT

A crucial component of nonalcoholic fatty liver disease (NAFLD) pathogenesis is lipid stress, which may contribute to hepatic inflammation and activation of innate immunity in the liver. However, little is known regarding how dietary lipids, including fat and cholesterol, may facilitate innate immune activation in vivo. We hypothesized that dietary fat and cholesterol drive NAFLD progression to steatohepatitis and hepatic fibrosis by altering the transcription and phenotype of hepatic macrophages. This hypothesis was tested by using RNA-sequencing methods to characterize and analyze sort-purified hepatic macrophage populations that were isolated from mice fed diets with varying amounts of fat and cholesterol. The addition of cholesterol to a high-fat diet triggered hepatic pathology reminiscent of advanced nonalcoholic steatohepatitis (NASH) in humans characterized by signs of cholesterol dysregulation, generation of oxidized low-density lipoprotein, increased recruitment of hepatic macrophages, and significant fibrosis. RNA-sequencing analyses of hepatic macrophages in this model revealed that dietary cholesterol induced a tissue repair and regeneration phenotype in Kupffer cells (KCs) and recruited infiltrating macrophages to a greater degree than fat. Furthermore, comparison of diseased KCs and infiltrating macrophages revealed that these two macrophage subsets are transcriptionally diverse. Finally, direct stimulation of murine and human macrophages with oxidized low-density lipoprotein recapitulated some of the transcriptional changes observed in the RNA-sequencing study. These findings indicate that fat and cholesterol synergize to alter macrophage phenotype, and they also challenge the dogma that KCs are purely proinflammatory in NASH. Conclusion: This comprehensive view of macrophage populations in NASH indicates mechanisms by which cholesterol contributes to NASH progression and identifies potential therapeutic targets for this common disease.


Subject(s)
Cholesterol, Dietary/adverse effects , Kupffer Cells/metabolism , Liver/immunology , Non-alcoholic Fatty Liver Disease/etiology , Animals , Disease Progression , Hepatitis/etiology , Kupffer Cells/ultrastructure , Lipid Metabolism , Liver/pathology , Male , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Transcriptome
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.
Hepatology ; 66(1): 271-279, 2017 07.
Article in English | MEDLINE | ID: mdl-28195343

ABSTRACT

Glycan-binding proteins, which include galectins, are involved at all stages of immunity and inflammation, from initiation through resolution. Galectin-9 (Gal-9) is highly expressed in the liver and has a wide variety of biological functions in innate and adaptive immunity that are instrumental in the maintenance of hepatic homeostasis. In the setting of viral hepatitis, increased expression of Gal-9 drives the expansion of regulatory T cells and contraction of effector T cells, thereby favoring viral persistence. The dichotomous nature of Gal-9 is evident in hepatocellular carcinoma, where loss of expression in hepatocytes promotes tumor growth and metastasis, whereas overexpression by Kupffer cells and endothelial cells inhibits the antitumor immune response. In nonalcoholic fatty liver disease, Gal-9 is involved indirectly in the expansion of protective natural killer T-cell populations. In ischemic liver injury, hepatocyte-derived Gal-9 is both diagnostic and cytoprotective. In drug-induced acute liver failure, plasma levels correlate with outcome. Here, we offer a synthesis of recent and emerging findings on Gal-9 in the regulation of hepatic inflammation. Ongoing studies are warranted to better elucidate the pathophysiology of hepatic immune-mediated diseases and to develop new therapeutic interventions using glycan-binding proteins. (Hepatology 2017;66:271-279).


Subject(s)
Adaptive Immunity/physiology , Galectins/metabolism , Homeostasis/immunology , Liver Diseases/immunology , Liver Diseases/physiopathology , Animals , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/physiopathology , Hepatitis/immunology , Hepatitis/physiopathology , Hepatitis, Autoimmune/immunology , Hepatitis, Autoimmune/physiopathology , Humans , Immunity, Innate/physiology , Liver Diseases, Alcoholic/immunology , Liver Diseases, Alcoholic/physiopathology , Liver Failure, Acute/immunology , Liver Failure, Acute/physiopathology , Liver Neoplasms/immunology , Liver Neoplasms/physiopathology , Sensitivity and Specificity
7.
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
8.
Liver Transpl ; 23(2): 155-165, 2017 02.
Article in English | MEDLINE | ID: mdl-28006875

ABSTRACT

The Model for End-Stage Liver Disease (MELD) score has reduced accuracy for liver transplantation (LT) wait-list mortality when MELD ≤ 20. Neutrophil-to-lymphocyte ratio (NLR) is a biomarker associated with systemic inflammation and may predict cirrhotic decompensation and death. We aimed to evaluate the prognostic utility of high NLR (≥4) for liver-related death among low MELD patients listed for LT, controlling for stage of cirrhosis. In a nested case-control study of cirrhotic adults awaiting LT (February 2002 to May 2011), cases were LT candidates with a liver-related death and MELD ≤ 20 within 90 days of death. Controls were similar LT candidates who were alive for ≥90 days after LT listing. NLR and other covariates were assessed at the date of lowest MELD, within 90 days of death for cases and within 90 days after listing for controls. There were 41 cases and 66 controls; MELD scores were similar. NLR 25th, 50th, 75th percentile cutoffs were 1.9, 3.1, and 6.8. NLR was ≥ 4 in 25/41 (61%) cases and in 17/66 (26%) controls. In univariate analysis, NLR (continuous ≥ 1.9, ≥ 4, ≥ 6.8), increasing cirrhosis stage, jaundice, encephalopathy, serum sodium, and albumin and nonselective beta-blocker use were significantly (P < 0.01) associated with liver-related death. In multivariate analysis, NLR of ≥1.9, ≥ 4, ≥ 6.8 were each associated with liver-related death. Furthermore, we found that NLR correlated with the frequency of circulating low-density granulocytes, previously identified as displaying proinflammatory properties, as well as monocytes. In conclusion, elevated NLR is associated with liver-related death, independent of MELD and cirrhosis stage. High NLR may aid in determining risk for cirrhotic decompensation, need for increased monitoring, and urgency for expedited LT in candidates with low MELD. Liver Transplantation 23 155-165 2017 AASLD.


Subject(s)
End Stage Liver Disease/mortality , Liver Cirrhosis/mortality , Liver Transplantation , Lymphocytes , Neutrophils , Waiting Lists/mortality , Biomarkers/blood , Case-Control Studies , End Stage Liver Disease/blood , End Stage Liver Disease/etiology , Female , Humans , Liver Cirrhosis/blood , Liver Cirrhosis/complications , Lymphocyte Count , Male , Middle Aged , Multivariate Analysis , Prognosis , Risk Factors , Severity of Illness Index
9.
J Immunol ; 195(8): 3737-47, 2015 10 15.
Article in English | MEDLINE | ID: mdl-26342030

ABSTRACT

Hepatitis C virus (HCV) is the world's most common blood-borne viral infection for which there is no vaccine. The rates of vertical transmission range between 3 and 6% with odds 90% higher in the presence of HIV coinfection. Prevention of vertical transmission is not possible because of lack of an approved therapy for use in pregnancy or an effective vaccine. Recently, HCV has been identified as an independent risk factor for preterm delivery, perinatal mortality, and other complications. In this study, we characterized the immune responses that contribute to the control of viral infection at the maternal-fetal interface (MFI) in the early gestational stages. In this study, we show that primary human trophoblast cells and an extravillous trophoblast cell line (HTR8), from first and second trimester of pregnancy, express receptors relevant for HCV binding/entry and are permissive for HCV uptake. We found that HCV-RNA sensing by human trophoblast cells induces robust upregulation of type I/III IFNs and secretion of multiple chemokines that elicit recruitment and activation of decidual NK cells. Furthermore, we observed that HCV-RNA transfection induces a proapoptotic response within HTR8 that could affect the morphology of the placenta. To our knowledge, for the first time, we demonstrate that HCV-RNA sensing by human trophoblast cells elicits a strong antiviral response that alters the recruitment and activation of innate immune cells at the MFI. This work provides a paradigm shift in our understanding of HCV-specific immunity at the MFI as well as novel insights into mechanisms that limit vertical transmission but may paradoxically lead to virus-related pregnancy complications.


Subject(s)
Hepacivirus/immunology , Hepatitis C/immunology , Immunity, Maternally-Acquired , Killer Cells, Natural/immunology , Pregnancy Complications, Infectious/immunology , Trophoblasts/immunology , Adult , Female , Hepatitis C/pathology , Hepatitis C/transmission , Humans , Immunity, Innate , Infectious Disease Transmission, Vertical , Killer Cells, Natural/pathology , Pregnancy , Pregnancy Complications, Infectious/virology , Trophoblasts/pathology
10.
Immunol Rev ; 255(1): 68-81, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23947348

ABSTRACT

Natural killer cells (NKs) are involved in every stage of hepatitis C viral (HCV) infection, from protection against HCV acquisition and resolution in the acute phase to treatment-induced clearance. In addition to their direct antiviral actions, NKs are involved in the induction and priming of appropriate downstream T-cell responses. In the setting of chronic HCV, overall NK cell levels are decreased, subset distribution is altered, and changes in NK receptor (NKR) expression have been demonstrated, although the contribution of individual NKRs to viral clearance or persistence remains to be clarified. Enhanced NK cell cytotoxicity accompanied by insufficient interferon-γ production may promote liver damage in the setting of chronic infection. Treatment-induced clearance is associated with activation of NK cells, and it will be of interest to monitor NK cell responses to triple therapy. Activated NK cells also have anti-fibrotic properties, and the same hepatic NK cell populations that are actively involved in control of HCV may also be involved in control of HCV-associated liver damage. We still have much to learn, in particular: how do liver-derived NKs influence the outcome of HCV infection? Do NK receptors recognize HCV-specific components? And, are HCV-specific memory NK populations generated?


Subject(s)
Hepacivirus/immunology , Hepatitis C/immunology , Killer Cells, Natural/immunology , Animals , Fibrosis/immunology , Fibrosis/metabolism , Hepatitis C/drug therapy , Hepatitis C/metabolism , Humans , Immunomodulation , Killer Cells, Natural/metabolism , Liver/immunology , Liver/metabolism , Liver/pathology , Liver/virology , Lymphocyte Activation/immunology , Phenotype
11.
Clin Gastroenterol Hepatol ; 14(5): 762-8.e1, 2016 May.
Article in English | MEDLINE | ID: mdl-26598225

ABSTRACT

BACKGROUND & AIMS: Alcohol consumption is a major cause of chronic liver disease and contributes to a large proportion of cirrhosis-related deaths worldwide. However, only a fraction of heavy consumers of alcohol develop advanced alcoholic liver disease (ALD), so there are likely to be other risk factors. We investigated whether polymorphisms in the gene encoding galectin-9 (LGALS9), previously shown to mediate liver injury, were associated with the development of ALD. METHODS: We isolated DNA from peripheral blood mononuclear cells (PBMCs) of 575 individuals with at-risk alcohol consumption but no other risk factors for chronic liver disease; all subjects were white Europeans who had consumed more than 80 grams ethanol per day. Of the subjects, 388 had ALD (including, 268 with cirrhosis and 74 with alcoholic hepatitis; mean age, 49 y; 72% male) and 187 had normal liver function with no biochemical or clinical evidence of liver disease (controls; mean age, 42 y; 73% male). Select LGALS9 polymorphisms were genotyped using allelic discrimination. We also genotyped and measured expression of LGALS9 messenger RNA in PBMCs from individuals who were not heavy consumers of alcohol. RESULTS: We used data from the HapMap project to identify 5 single-nucleotide polymorphisms (SNPs) that tag all the common haplotypes. When we looked for these SNPs in individuals with vs without liver disease, 4 (rs3751093, rs4239242, rs732222, and rs4794976) were associated with an increased risk of developing ALD. We found that levels of LGALS9 messenger RNA and protein expressed were associated with an allele carried by PBMCs. Multivariate analysis confirmed that rs4239242 and rs4794976 were associated with an increased risk of ALD. CONCLUSIONS: In a genetic analysis of heavy consumers of alcohol, we associated 2 SNPS in LGALS9 with the development of ALD. Although larger studies are required, this information could be used to determine the risk of individuals developing ALD or to develop therapeutic agents.


Subject(s)
Alcoholism/complications , Galectins/genetics , Genetic Predisposition to Disease , Liver Cirrhosis, Alcoholic/genetics , Polymorphism, Single Nucleotide , Galectins/biosynthesis , Gene Expression Profiling , Genotype , RNA, Messenger/analysis
12.
Gastroenterology ; 148(2): 392-402.e13, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25447848

ABSTRACT

BACKGROUND & AIMS: Liver sinusoidal endothelial cells (LSECs) make up a large proportion of the nonparenchymal cells in the liver. LSECs are involved in induction of immune tolerance, but little is known about their functions during hepatitis C virus (HCV) infection. METHODS: Primary human LSECs (HLSECs) and immortalized liver endothelial cells (TMNK-1) were exposed to various forms of HCV, including full-length transmitted/founder virus, sucrose-purified Japanese fulminant hepatitis-1 (JFH-1), a virus encoding a luciferase reporter, and the HCV-specific pathogen-associated molecular pattern molecules. Cells were analyzed by confocal immunofluorescence, immunohistochemical, and polymerase chain reaction assays. RESULTS: HLSECs internalized HCV, independent of cell-cell contacts; HCV RNA was translated but not replicated. Through pattern recognition receptors (Toll-like receptor 7 and retinoic acid-inducible gene 1), HCV RNA induced consistent and broad transcription of multiple interferons (IFNs); supernatants from primary HLSECs transfected with HCV-specific pathogen-associated molecular pattern molecules increased induction of IFNs and IFN-stimulated genes in HLSECs. Recombinant type I and type III IFNs strongly up-regulated HLSEC transcription of IFN λ3 (IFNL3) and viperin (RSAD2), which inhibit replication of HCV. Compared with CD8(+) T cells, HLSECs suppressed HCV replication within Huh7.5.1 cells, also inducing IFN-stimulated genes in co-culture. Conditioned media from IFN-stimulated HLSECs induced expression of antiviral genes by uninfected primary human hepatocytes. Exosomes, derived from HLSECs after stimulation with either type I or type III IFNs, controlled HCV replication in a dose-dependent manner. CONCLUSIONS: Cultured HLSECs produce factors that mediate immunity against HCV. HLSECs induce self-amplifying IFN-mediated responses and release of exosomes with antiviral activity.


Subject(s)
Autocrine Communication , Endothelial Cells/physiology , Exosomes/physiology , Hepacivirus/physiology , Interferons/biosynthesis , Liver/cytology , Virus Replication , Cells, Cultured , Clathrin/physiology , Endothelial Cells/virology , Flow Cytometry , Hepatocytes/virology , Humans , Immunity, Innate , Interleukins/genetics
13.
J Pediatr ; 170: 142-8.e1, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26743497

ABSTRACT

OBJECTIVES: To assess if peripheral T cell populations in children with chronic hepatitis C virus (HCV) infection would show evidence of activation/exhaustion and an attenuated functional response. STUDY DESIGN: Compared with adults, children with HCV infection have a higher rate of spontaneous viral clearance. In adults, chronic HCV has been linked to T cell exhaustion. Little is known of the immune status of children with HCV. Peripheral blood mononuclear cells were isolated from 16 children with HCV (6 males, 10 females; mean age 8.6 years, range 2-17), 16 age- and sex-matched control children without HCV infection, and 20 adults with chronic HCV. Multiparameter flow cytometry was performed to characterize T cell differences across the 3 groups. RESULTS: Controls and children with HCV had similar levels of CD4(+), CD8(+), and γδ(+) T cells. Children with HCV demonstrated a decrease in naïve T cells compared with control children and increased activation/exhaustion marker expression on both CD8(+) and CD4(+) T cells. Transcription factor analysis suggested functional activation of T cells in children with HCV; however, only the CD4(+) subset had enhanced cytokine production (interferon gamma and interleukin-2) compared with control children. CONCLUSIONS: The HCV response in children is characterized by several changes in T cell phenotype. Many of these changes, such as increased T cell expression of programmed cell death-1, are similar to responses in adults. Of note, cytokine production by CD4(+) helper T cells is increased in children with HCV compared with age- and sex-matched control children, which may influence long-term prognosis in children with HCV.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Hepatitis C, Chronic/blood , Adolescent , Adult , Case-Control Studies , Cells, Cultured , Child , Child, Preschool , Female , Flow Cytometry , Hepatitis C, Chronic/immunology , Humans , Interferon-gamma/metabolism , Interleukin-2/metabolism , Lymphocyte Activation , Male , Programmed Cell Death 1 Receptor/metabolism , Receptors, Immunologic/metabolism , T-Box Domain Proteins/metabolism
14.
Clin Immunol ; 158(1): 114-25, 2015 May.
Article in English | MEDLINE | ID: mdl-25797693

ABSTRACT

T cell immunoglobulin- and mucin-domain-containing molecule-3 (Tim-3) is an inhibitory receptor implicated in T cell exhaustion characteristic of chronic viral infection. Limited data exist on NK cell Tim-3 expression and functional consequences. In chronic hepatitis C virus (HCV)-infected subjects, we found increased Tim-3 on NKs, which was associated with an activated phenotype. The high level of Tim-3 was not reversed by successful IFN-alpha-based antiviral therapy. Tim-3(high) NK cells up-regulated TRAIL in response to IFN-alpha to a greater extent and demonstrated greater lymphokine-activated killing activity, viral control, and degranulation but similar cytokine production than their Tim-3(low) counterparts. Our results suggest that Tim-3 on NKs is associated with activation of this innate lymphocyte population that is polarized towards cytotoxicity in chronic HCV. These findings reveal roles for Tim-3 in the regulation of NKs that might represent targets for treatment of chronic viral infections.


Subject(s)
Cytotoxicity, Immunologic/immunology , Hepatitis C, Chronic/immunology , Killer Cells, Natural/immunology , Membrane Proteins/immunology , Adult , Aged , Antiviral Agents/pharmacology , Case-Control Studies , Cytokines/immunology , Cytotoxicity, Immunologic/drug effects , Female , Hepatitis A Virus Cellular Receptor 2 , Humans , Interferon-alpha/pharmacology , Killer Cells, Natural/drug effects , Male , Membrane Proteins/drug effects , Middle Aged , TNF-Related Apoptosis-Inducing Ligand/metabolism , Up-Regulation , Young Adult
15.
PLoS Pathog ; 9(4): e1003316, 2013.
Article in English | MEDLINE | ID: mdl-23637605

ABSTRACT

Plasmacytoid Dendritic Cells (pDCs) represent a key immune cell in the defense against viruses. Through pattern recognition receptors (PRRs), these cells detect viral pathogen associated molecular patterns (PAMPs) and initiate an Interferon (IFN) response. pDCs produce the antiviral IFNs including the well-studied Type I and the more recently described Type III. Recent genome wide association studies (GWAS) have implicated Type III IFNs in HCV clearance. We examined the IFN response induced in a pDC cell line and ex vivo human pDCs by a region of the HCV genome referred to as the HCV PAMP. This RNA has been shown previously to be immunogenic in hepatocytes, whereas the conserved X-region RNA is not. We show that in response to the HCV PAMP, pDC-GEN2.2 cells upregulate and secrete Type III (in addition to Type I) IFNs and upregulate PRR genes and proteins. We also demonstrate that the recognition of this RNA is dependent on RIG-I-like Receptors (RLRs) and Toll-like Receptors (TLRs), challenging the dogma that RLRs are dispensable in pDCs. The IFNs produced by these cells in response to the HCV PAMP also control HCV replication in vitro. These data are recapitulated in ex vivo pDCs isolated from healthy donors. Together, our data shows that pDCs respond robustly to HCV RNA to make Type III Interferons that control viral replication. This may represent a novel therapeutic strategy for the treatment of HCV.


Subject(s)
Dendritic Cells/immunology , Hepacivirus/immunology , Hepatitis C/immunology , Interferon Type I/biosynthesis , Cell Line, Tumor , Culture Media, Conditioned , Dendritic Cells/metabolism , Humans , Interferons , Interleukins/biosynthesis , RNA Interference , RNA, Small Interfering , RNA, Viral/immunology , Receptors, Retinoic Acid , Toll-Like Receptors/metabolism , Virus Replication
16.
J Mol Cell Cardiol ; 67: 112-25, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24374140

ABSTRACT

Fibrosis, which is defined as excessive accumulation of fibrous connective tissue, contributes to the pathogenesis of numerous diseases involving diverse organ systems. Cardiac fibrosis predisposes individuals to myocardial ischemia, arrhythmias and sudden death, and is commonly associated with diastolic dysfunction. Histone deacetylase (HDAC) inhibitors block cardiac fibrosis in pre-clinical models of heart failure. However, which HDAC isoforms govern cardiac fibrosis, and the mechanisms by which they do so, remains unclear. Here, we show that selective inhibition of class I HDACs potently suppresses angiotensin II (Ang II)-mediated cardiac fibrosis by targeting two key effector cell populations, cardiac fibroblasts and bone marrow-derived fibrocytes. Class I HDAC inhibition blocks cardiac fibroblast cell cycle progression through derepression of the genes encoding the cyclin-dependent kinase (CDK) inhibitors, p15 and p57. In contrast, class I HDAC inhibitors block agonist-dependent differentiation of fibrocytes through a mechanism involving repression of ERK1/2 signaling. These findings define novel roles for class I HDACs in the control of pathological cardiac fibrosis. Furthermore, since fibrocytes have been implicated in the pathogenesis of a variety of human diseases, including heart, lung and kidney failure, our results suggest broad utility for isoform-selective HDAC inhibitors as anti-fibrotic agents that function, in part, by targeting these circulating mesenchymal cells.


Subject(s)
Angiotensin II/metabolism , Fibroblasts/drug effects , Fibrosis/physiopathology , Histone Deacetylase Inhibitors/pharmacology , Animals , Cell Cycle/drug effects , Cell Differentiation , Fibroblasts/metabolism , Fibrosis/drug therapy , Flow Cytometry , Humans , Immunoblotting , Male , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Protein Isoforms/pharmacology
17.
J Biol Chem ; 288(17): 11761-70, 2013 Apr 26.
Article in English | MEDLINE | ID: mdl-23460643

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) affects a large proportion of the American population. The spectrum of disease ranges from bland steatosis without inflammation to nonalcoholic steatohepatitis and cirrhosis. Bile acids are critical regulators of hepatic lipid and glucose metabolism and signal through two major receptor pathways: farnesoid X receptor (FXR), a member of the nuclear hormone receptor superfamily, and TGR5, a G protein-coupled bile acid receptor (GPBAR1). Both FXR and TGR5 demonstrate pleiotropic functions, including immune modulation. To evaluate the effects of these pathways in NAFLD, we treated obese db/db mice with a dual FXR/TGR5 agonist (INT-767) for 6 weeks. Treatment with the agonist significantly improved the histological features of nonalcoholic steatohepatitis. Furthermore, treatment increased the proportion of intrahepatic monocytes with the anti-inflammatory Ly6C(low) phenotype and increased intrahepatic expression of genes expressed by alternatively activated macrophages, including CD206, Retnla, and Clec7a. In vitro treatment of monocytes with INT-767 led to decreased Ly6C expression and increased IL-10 production through a cAMP-dependent pathway. Our data indicate that FXR/TGR5 activation coordinates the immune phenotype of monocytes and macrophages, both in vitro and in vivo, identifying potential targeting strategies for treatment of NAFLD.


Subject(s)
Fatty Liver/metabolism , Liver/metabolism , Monocytes/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cyclic AMP/immunology , Cyclic AMP/metabolism , Fatty Liver/immunology , Fatty Liver/pathology , Gene Expression Regulation/immunology , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/immunology , Interleukin-10/biosynthesis , Interleukin-10/immunology , Lectins, C-Type/biosynthesis , Lectins, C-Type/immunology , Liver/immunology , Liver/pathology , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Male , Mannose Receptor , Mannose-Binding Lectins/biosynthesis , Mannose-Binding Lectins/immunology , Mice , Mice, Obese , Monocytes/immunology , Monocytes/pathology , Non-alcoholic Fatty Liver Disease , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/immunology , Receptors, Cytoplasmic and Nuclear/immunology , Receptors, G-Protein-Coupled/immunology
19.
J Virol ; 87(9): 4835-45, 2013 May.
Article in English | MEDLINE | ID: mdl-23408620

ABSTRACT

Galectin-9 is a pleiotropic immune modulator affecting numerous cell types of innate and adaptive immunity. Patients with chronic infection with either hepatitis C virus (HCV) or HIV have elevated circulating levels. Limited data exist on the regulation of natural killer (NK) cell function through interaction with galectin-9. We found that galectin-9 ligation downregulates multiple immune-activating genes, including eight involved in the NK cell-mediated cytotoxicity pathway, impairs lymphokine-activated killing, and decreases the proportion of gamma interferon (IFN-γ)-producing NK cells that had been stimulated with interleukin-12 (IL-12)/IL-15. We demonstrate that the transcriptional and functional changes induced by galectin-9 are independent of Tim-3. Consistent with these results for humans, we find that the genetic absence of galectin-9 in mice is associated with greater IFN-γ production by NK cells and enhanced degranulation. We also show that in the setting of a short-term (4-day) murine cytomegalovirus infection, terminally differentiated NKs accumulate in the livers of galectin-9 knockout mice, and that hepatic NKs spontaneously produce significantly more IFN-γ in this setting. Taken together, our results indicate that galectin-9 engagement impairs the function of NK cells, including cytotoxicity and cytokine production.


Subject(s)
Galectins/immunology , Herpesviridae Infections/immunology , Killer Cells, Natural/immunology , Animals , Cells, Cultured , Cytotoxicity, Immunologic , Galectins/genetics , Hepatitis A Virus Cellular Receptor 2 , Herpesviridae Infections/genetics , Herpesviridae Infections/virology , Humans , Interleukin-12/immunology , Interleukin-15/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Muromegalovirus/physiology , Receptors, Virus/genetics , Receptors, Virus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL