Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Cell ; 180(3): 490-501.e16, 2020 02 06.
Article in English | MEDLINE | ID: mdl-31955848

ABSTRACT

Integrin αvß8 binds with exquisite specificity to latent transforming growth factor-ß (L-TGF-ß). This binding is essential for activating L-TGF-ß presented by a variety of cell types. Inhibiting αvß8-mediated TGF-ß activation blocks immunosuppressive regulatory T cell differentiation, which is a potential therapeutic strategy in cancer. Using cryo-electron microscopy, structure-guided mutagenesis, and cell-based assays, we reveal the binding interactions between the entire αvß8 ectodomain and its intact natural ligand, L-TGF-ß, as well as two different inhibitory antibody fragments to understand the structural underpinnings of αvß8 binding specificity and TGF-ß activation. Our studies reveal a mechanism of TGF-ß activation where mature TGF-ß signals within the confines of L-TGF-ß and the release and diffusion of TGF-ß are not required. The structural details of this mechanism provide a rational basis for therapeutic strategies to inhibit αvß8-mediated L-TGF-ß activation.


Subject(s)
Cryoelectron Microscopy/methods , Integrins/chemistry , Integrins/metabolism , Latent TGF-beta Binding Proteins/chemistry , Latent TGF-beta Binding Proteins/metabolism , Transforming Growth Factor beta1/chemistry , Transforming Growth Factor beta1/metabolism , Animals , Antibodies/immunology , Binding Sites , Bronchi/cytology , CHO Cells , Cricetulus , Female , Humans , Immunoglobulin Fab Fragments/immunology , Integrins/immunology , Lymphocyte Activation , Male , Mink , Protein Binding , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , T-Lymphocytes, Regulatory/immunology
2.
Am J Physiol Lung Cell Mol Physiol ; 312(1): L122-L130, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27913421

ABSTRACT

Small airway fibrosis is a major pathological feature of chronic obstructive pulmonary disease (COPD) and is refractory to current treatments. Chronic inflammatory cells accumulate around small airways in COPD and are thought to play a major role in small airway fibrosis. Mice deficient in α/ß T cells have recently been shown to be protected from both experimental airway inflammation and fibrosis. In these models, CD4+Th17 cells and secretion of IL-17A are increased. However, a pathogenic role for IL-17 in specifically mediating fibrosis around airways has not been demonstrated. Here a role for IL-17A in airway fibrosis was demonstrated using mice deficient in the IL-17 receptor A (il17ra) Il17ra-deficient mice were protected from both airway inflammation and fibrosis in two different models of airway fibrosis that employ COPD-relevant stimuli. In these models, CD4+ Th17 are a major source of IL-17A with other expressing cell types including γδ T cells, type 3 innate lymphoid cells, polymorphonuclear cells, and CD8+ T cells. Antibody neutralization of IL-17RA or IL-17A confirmed that IL-17A was the relevant pathogenic IL-17 isoform and IL-17RA was the relevant receptor in airway inflammation and fibrosis. These results demonstrate that the IL-17A/IL-17 RA axis is crucial to murine airway fibrosis. These findings suggest that IL-17 might be targeted to prevent the progression of airway fibrosis in COPD.


Subject(s)
Interleukin-17/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Adenoviridae/metabolism , Animals , Disease Models, Animal , Interleukin-1beta/pharmacology , Mice, Inbred C57BL , Neutralization Tests , Pneumonia/complications , Pneumonia/metabolism , Pneumonia/pathology , Poly I-C/pharmacology , Pulmonary Disease, Chronic Obstructive/complications , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Fibrosis/complications , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Receptors, Interleukin-17/metabolism , Smoking/adverse effects
3.
J Immunol ; 194(8): 3962-9, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25786688

ABSTRACT

Chronic airway inflammation and fibrosis, known as airway remodeling, are defining features of chronic obstructive pulmonary disease and are refractory to current treatments. How and whether chronic inflammation contributes to airway fibrosis remain controversial. In this study, we use a model of chronic obstructive pulmonary disease airway disease utilizing adenoviral delivery of IL-1ß to determine that adaptive T cell immunity is required for airway remodeling because mice deficient in α/ß T cells (tcra(-/-)) are protected. Dendritic cells (DCs) accumulate around chronic obstructive pulmonary disease airways and are critical to prime adaptive immunity, but they have not been shown to directly influence airway remodeling. We show that DC depletion or deficiency in the crucial DC chemokine receptor ccr6 both protect from adenoviral IL-1ß-induced airway adaptive T cell immune responses and fibrosis in mice. These results provide evidence that chronic airway inflammation, mediated by accumulation of α/ß T cells and driven by DCs, is critical to airway fibrosis.


Subject(s)
Adaptive Immunity , Dendritic Cells/immunology , Interleukin-1beta/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Fibrosis/immunology , Animals , Dendritic Cells/pathology , Interleukin-1beta/genetics , Mice , Mice, Knockout , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Fibrosis/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
4.
J Immunol ; 195(3): 1182-90, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26109638

ABSTRACT

Small airway chronic inflammation is a major pathologic feature of chronic obstructive pulmonary disease (COPD) and is refractory to current treatments. Dendritic cells (DCs) accumulate around small airways in COPD. DCs are critical mediators of Ag surveillance and Ag presentation and amplify adaptive immune responses. How DCs accumulate around airways remains largely unknown. We use 2-photon DC imaging of living murine lung sections to directly visualize the dynamic movement of living DCs around airways in response to either soluble mediators (IL-1ß) or environmental stimuli (cigarette smoke or TLR3 ligands) implicated in COPD pathogenesis. We find that DCs accumulate around murine airways primarily by increasing velocity (chemokinesis) rather than directional migration (chemotaxis) in response to all three stimuli. DC accumulation maximally occurs in a specific zone located 26-50 µm from small airways, which overlaps with zones of maximal DC velocity. Our data suggest that increased accumulation of DCs around airways results from increased numbers of highly chemokinetic DCs entering the lung from the circulation with balanced rates of immigration and emigration. Increases in DC accumulation and chemokinesis are partially dependent on ccr6, a crucial DC chemokine receptor, and fibroblast expression of the integrin αvß8, a critical activator of TGF-ß. αvß8-Mediated TGF-ß activation is known to enhance IL-1ß-dependent fibroblast expression of the only known endogenous ccr6 chemokine ligand, ccl20. Taken together, these data suggest a mechanism by which αvß8, ccl20, and ccr6 interact to lead to DC accumulation around airways in response to COPD-relevant stimuli.


Subject(s)
Dendritic Cells/immunology , Integrins/immunology , Interleukin-1beta/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Transforming Growth Factor beta/immunology , Adaptive Immunity/immunology , Animals , Cell Movement/immunology , Chemokine CCL20/biosynthesis , Chemokine CCL20/immunology , Disease Models, Animal , Enzyme Activation/immunology , Fibroblasts/immunology , Integrins/biosynthesis , Interleukin-1beta/biosynthesis , Lung/diagnostic imaging , Mice , Mice, Inbred C57BL , Mice, Knockout , Poly I-C/pharmacology , Pulmonary Disease, Chronic Obstructive/pathology , Radiography , Receptors, CCR6/genetics , Receptors, CCR6/immunology , Smoke/adverse effects , Toll-Like Receptor 3 , Transforming Growth Factor beta/metabolism
5.
J Biol Chem ; 290(23): 14717-28, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-25918170

ABSTRACT

CCL20 is the only chemokine ligand for the chemokine receptor CCR6, which is expressed by the critical antigen presenting cells, dendritic cells. Increased expression of CCL20 is likely involved in the increased recruitment of dendritic cells observed in fibroinflammatory diseases such as chronic obstructive pulmonary disease (COPD). CCL20 expression is increased by the proinflammatory cytokine IL-1ß. We have determined that IL-1ß-dependent CCL20 expression is also dependent on the multifunctional cytokine TGF-ß. TGF-ß is expressed in a latent form that must be activated to function, and activation is achieved through binding to the integrin αvß8 (itgb8). Here we confirm correlative increases in αvß8 and IL-1ß with CCL20 protein in lung parenchymal lysates of a large cohort of COPD patients. How IL-1ß- and αvß8-mediated TGF-ß activation conspire to increase fibroblast CCL20 expression remains unknown, because these pathways have not been shown to directly interact. We evaluate the 5'-flanking region of CCL20 to determine that IL-1ß-driven CCL20 expression is dependent on αvß8-mediated activation of TGF-ß. We identify a TGF-ß-responsive element (i.e. SMAD) located on an upstream enhancer of the human CCL20 promoter required for efficient IL-1ß-dependent CCL20 expression. By chromatin immunoprecipitation, this upstream enhancer complexes with the p50 subunit of NF-κB on a NF-κB-binding element close to the transcriptional start site of CCL20. These interactions are confirmed by electromobility shift assays in nuclear extracts from human lung fibroblasts. These data define a mechanism by which αvß8-dependent activation of TGF-ß regulates IL-1ß-dependent CCL20 expression in COPD.


Subject(s)
Chemokine CCL20/genetics , Interleukin-1beta/immunology , Response Elements , Signal Transduction , Transforming Growth Factor beta/immunology , Animals , Base Sequence , Cells, Cultured , Fibroblasts/immunology , Fibroblasts/metabolism , Gene Expression Regulation , Humans , Lung/cytology , Mice , Mice, Inbred C57BL , NF-kappa B/immunology
6.
bioRxiv ; 2023 Mar 11.
Article in English | MEDLINE | ID: mdl-36945648

ABSTRACT

In the past decade, high-dimensional single cell technologies have revolutionized basic and translational immunology research and are now a key element of the toolbox used by scientists to study the immune system. However, analysis of the data generated by these approaches often requires clustering algorithms and dimensionality reduction representation which are computationally intense and difficult to evaluate and optimize. Here we present Cyclone, an analysis pipeline integrating dimensionality reduction, clustering, evaluation and optimization of clustering resolution, and downstream visualization tools facilitating the analysis of a wide range of cytometry data. We benchmarked and validated Cyclone on mass cytometry (CyTOF), full spectrum fluorescence-based cytometry, and multiplexed immunofluorescence (IF) in a variety of biological contexts, including infectious diseases and cancer. In each instance, Cyclone not only recapitulates gold standard immune cell identification, but also enables the unsupervised identification of lymphocytes and mononuclear phagocytes subsets that are associated with distinct biological features. Altogether, the Cyclone pipeline is a versatile and accessible pipeline for performing, optimizing, and evaluating clustering on variety of cytometry datasets which will further power immunology research and provide a scaffold for biological discovery.

7.
mBio ; : e0131823, 2023 Nov 08.
Article in English | MEDLINE | ID: mdl-37938000

ABSTRACT

Hepatitis C virus (HCV) is the leading cause of death from liver disease. How HCV infection causes lasting liver damage and increases cancer risk remains unclear. Here, we identify bipotent liver stem cells as novel targets for HCV infection, and their erroneous differentiation as the potential cause of impaired liver regeneration and cancer development. We show 3D organoids generated from liver stem cells from actively HCV-infected individuals carry replicating virus and maintain low-grade infection over months. Organoids can be infected with a primary HCV isolate. Virus-inclusive single-cell RNA sequencing uncovered transcriptional reprogramming in HCV+ cells supporting hepatocytic differentiation, cancer stem cell development, and viral replication while stem cell proliferation and interferon signaling are disrupted. Our data add a new pathogenesis mechanism-infection of liver stem cells-to the biology of HCV infection that may explain progressive liver damage and enhanced cancer risk through an altered stem cell state.ImportanceThe hepatitis C virus (HCV) causes liver disease, affecting millions. Even though we have effective antivirals that cure HCV, they cannot stop terminal liver disease. We used an adult stem cell-derived liver organoid system to understand how HCV infection leads to the progression of terminal liver disease. Here, we show that HCV maintains low-grade infections in liver organoids for the first time. HCV infection in liver organoids leads to transcriptional reprogramming causing cancer cell development and altered immune response. Our finding shows how HCV infection in liver organoids mimics HCV infection and patient pathogenesis. These results reveal that HCV infection in liver organoids contributes to liver disease progression.

8.
Front Immunol ; 14: 1167241, 2023.
Article in English | MEDLINE | ID: mdl-37731497

ABSTRACT

In the past decade, high-dimensional single-cell technologies have revolutionized basic and translational immunology research and are now a key element of the toolbox used by scientists to study the immune system. However, analysis of the data generated by these approaches often requires clustering algorithms and dimensionality reduction representation, which are computationally intense and difficult to evaluate and optimize. Here, we present Cytometry Clustering Optimization and Evaluation (Cyclone), an analysis pipeline integrating dimensionality reduction, clustering, evaluation, and optimization of clustering resolution, and downstream visualization tools facilitating the analysis of a wide range of cytometry data. We benchmarked and validated Cyclone on mass cytometry (CyTOF), full-spectrum fluorescence-based cytometry, and multiplexed immunofluorescence (IF) in a variety of biological contexts, including infectious diseases and cancer. In each instance, Cyclone not only recapitulates gold standard immune cell identification but also enables the unsupervised identification of lymphocytes and mononuclear phagocyte subsets that are associated with distinct biological features. Altogether, the Cyclone pipeline is a versatile and accessible pipeline for performing, optimizing, and evaluating clustering on a variety of cytometry datasets, which will further power immunology research and provide a scaffold for biological discovery.


Subject(s)
Cyclonic Storms , Algorithms , Benchmarking , Cluster Analysis , Technology
10.
Open Biol ; 12(3): 210320, 2022 03.
Article in English | MEDLINE | ID: mdl-35232252

ABSTRACT

Hepatitis C virus (HCV) remains a global public health challenge with an estimated 71 million people chronically infected, with surges in new cases and no effective vaccine. New methods are needed to study the human immune response to HCV since in vivo animal models are limited and in vitro cancer cell models often show dysregulated immune and proliferative responses. Here, we developed a CD8+ T cell and adult stem cell liver organoid system using a microfluidic chip to coculture 3D human liver organoids embedded in extracellular matrix with HLA-matched primary human T cells in suspension. We then employed automated phase contrast and immunofluorescence imaging to monitor T cell invasion and morphological changes in the liver organoids. This microfluidic coculture system supports targeted killing of liver organoids when pulsed with a peptide specific for HCV non-structural protein 3 (NS3) (KLVALGINAV) in the presence of patient-derived CD8+ T cells specific for KLVALGINAV. This demonstrates the novel potential of the coculture system to molecularly study adaptive immune responses to HCV in an in vitro setting using primary human cells.


Subject(s)
CD8-Positive T-Lymphocytes , Hepatitis C , Organoids , CD8-Positive T-Lymphocytes/immunology , Coculture Techniques , Hepacivirus , Hepatitis C/immunology , Humans , Microfluidics , Viral Nonstructural Proteins/immunology
11.
Sci Immunol ; 6(57)2021 03 26.
Article in English | MEDLINE | ID: mdl-33771888

ABSTRACT

Regulatory T cells (Tregs) that promote tumor immune evasion are enriched in certain tumors and correlate with poor prognosis. However, mechanisms for Treg enrichment remain incompletely understood. We described a mechanism for Treg enrichment in mouse and human tumors mediated by the αvß8 integrin. Tumor cell αvß8 bound to latent transforming growth factor-ß (L-TGF-ß) presented on the surface of T cells, resulting in TGF-ß activation and immunosuppressive Treg differentiation in vitro. In vivo, tumor cell αvß8 expression correlated with Treg enrichment, immunosuppressive Treg gene expression, and increased tumor growth, which was reduced in mice by αvß8 inhibition or Treg depletion. Structural modeling and cell-based studies suggested a highly geometrically constrained complex forming between αvß8-expressing tumor cells and L-TGF-ß-expressing T cells, facilitating TGF-ß activation, independent of release and diffusion, and providing limited access to TGF-ß inhibitors. These findings suggest a highly localized tumor-specific mechanism for Treg enrichment.


Subject(s)
Integrins/metabolism , Neoplasms/immunology , Neoplasms/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Tumor Escape , Animals , Biomarkers , Cell Line, Tumor , Computational Biology/methods , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Gene Expression Profiling , Humans , Mice , Models, Biological , Neoplasms/genetics , Neoplasms/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transcriptome
12.
J Exp Med ; 198(7): 1069-76, 2003 Oct 06.
Article in English | MEDLINE | ID: mdl-14530376

ABSTRACT

Natural killer (NK) and NK T cells are tissue lymphocytes that secrete cytokines rapidly upon stimulation. Here, we show that these cells maintain distinct patterns of constitutive cytokine mRNAs. Unlike conventional T cells, NK T cells activate interleukin (IL)-4 and interferon (IFN)-gamma transcription during thymic development and populate the periphery with both cytokine loci previously modified by histone acetylation. Similarly, NK cells transcribe and modify the IFN-gamma gene, but not IL-4, during developmental maturation in the bone marrow. Lineage-specific patterns of cytokine transcripts predate infection and suggest evolutionary selection for invariant but distinct types of effector responses among the earliest responding lymphocytes.


Subject(s)
Interferon-gamma/genetics , Interleukin-4/genetics , Killer Cells, Natural/immunology , RNA, Messenger/analysis , T-Lymphocytes/immunology , Animals , Chromatin/metabolism , Fluorescence , Killer Cells, Natural/physiology , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , T-Lymphocytes/physiology , Transcription, Genetic
13.
Proc Natl Acad Sci U S A ; 104(46): 18187-92, 2007 Nov 13.
Article in English | MEDLINE | ID: mdl-17991774

ABSTRACT

Hepatitis B virus (HBV) is a hepadnavirus that is a major cause of acute and chronic hepatitis in humans. Hepatitis B viral infection itself is noncytopathic, and it is the immune response to the viral antigens that is thought to be responsible for hepatic pathology. Previously, we developed a transgenic mouse model of primary HBV infection and demonstrated that the acute liver injury is mediated by nonclassical natural killer (NK)T cells, which are CD1d-restricted, but nonreactive to alpha-GalCer. We now demonstrate a role for NKG2D and its ligands in this nonclassical NKT cell-mediated immune response to hepatitis B virus and in the subsequent acute hepatitis that ensues. Surface expression of NKG2D and one of its ligands (retinoic acid early inducible-1 or RAE-1) are modulated in an HBV-dependent manner. Furthermore, blockade of an NKG2D-ligand interaction completely prevents the HBV- and CD1d-dependent, nonclassical NKT cell-mediated acute hepatitis and liver injury. This study has major implications for understanding activation of NKT cells and identifies a potential therapeutic target in treating hepatitis B viral infection.


Subject(s)
Hepatitis B virus/immunology , Hepatitis B/prevention & control , Killer Cells, Natural/immunology , Receptors, Immunologic/antagonists & inhibitors , Animals , Cell Line , Hepatitis B virus/physiology , Humans , Mice , Mice, Transgenic , NK Cell Lectin-Like Receptor Subfamily K , Receptors, Natural Killer Cell , Virus Replication
14.
J Cell Biol ; 157(3): 493-507, 2002 Apr 29.
Article in English | MEDLINE | ID: mdl-11970960

ABSTRACT

Integrins, matrix metalloproteases (MMPs), and the cytokine TGF-beta have each been implicated in homeostatic cell behaviors such as cell growth and matrix remodeling. TGF-beta exists mainly in a latent state, and a major point of homeostatic control is the activation of TGF-beta. Because the latent domain of TGF-beta1 possesses an integrin binding motif (RGD), integrins have the potential to sequester latent TGF-beta (SLC) to the cell surface where TGF-beta activation could be locally controlled. Here, we show that SLC binds to alpha(v)beta8, an integrin expressed by normal epithelial and neuronal cells in vivo. This binding results in the membrane type 1 (MT1)-MMP-dependent release of active TGF-beta, which leads to autocrine and paracrine effects on cell growth and matrix production. These data elucidate a novel mechanism of cellular homeostasis achieved through the coordination of the activities of members of three major gene families involved in cell-matrix interactions.


Subject(s)
Integrins/metabolism , Metalloendopeptidases/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cell Division/physiology , Cell Line , Homeostasis , Humans , Lung Neoplasms , Matrix Metalloproteinase 14 , Matrix Metalloproteinases, Membrane-Associated , Mice , Mice, Inbred BALB C , Oligopeptides/metabolism , Protein Binding , Protein Structure, Tertiary , Proteins/metabolism , TNF Receptor-Associated Factor 2 , Transfection , Transforming Growth Factor beta1 , Tumor Cells, Cultured
15.
Nat Struct Mol Biol ; 25(8): 698-704, 2018 08.
Article in English | MEDLINE | ID: mdl-30061598

ABSTRACT

Integrins are conformationally flexible cell surface receptors that survey the extracellular environment for their cognate ligands. Interactions with ligands are thought to be linked to global structural rearrangements involving transitions between bent, extended-closed and extended-open forms. Thus far, structural details are lacking for integrins in the extended conformations due to extensive flexibility between the headpiece and legs in this conformation. Here we present single-particle electron cryomicroscopy structures of human αvß8 integrin in the extended-closed conformation, which has been considered to be a low-affinity intermediate. Our structures show the headpiece rotating about a flexible αv knee, suggesting a ligand surveillance mechanism for integrins in their extended-closed form. Our model predicts that the extended conformation is mainly stabilized by an interface formed between flexible loops in the upper and lower domains of the αv leg. Confirming these findings with the αvß3 integrin suggests that our model of stabilizing the extended-closed conformation is generalizable to other integrins.


Subject(s)
Cryoelectron Microscopy/methods , Integrins/metabolism , Amino Acid Sequence , Humans , Integrins/chemistry , Protein Conformation , Sequence Homology, Amino Acid
16.
JCI Insight ; 3(20)2018 10 18.
Article in English | MEDLINE | ID: mdl-30333313

ABSTRACT

TGF-ß is a promising immunotherapeutic target. It is expressed ubiquitously in a latent form that must be activated to function. Determination of where and how latent TGF-ß (L-TGF-ß) is activated in the tumor microenvironment could facilitate cell- and mechanism-specific approaches to immunotherapeutically target TGF-ß. Binding of L-TGF-ß to integrin αvß8 results in activation of TGF-ß. We engineered and used αvß8 antibodies optimized for blocking or detection, which - respectively - inhibit tumor growth in syngeneic tumor models or sensitively and specifically detect ß8 in human tumors. Inhibition of αvß8 potentiates cytotoxic T cell responses and recruitment of immune cells to tumor centers - effects that are independent of PD-1/PD-L1. ß8 is expressed on the cell surface at high levels by tumor cells, not immune cells, while the reverse is true of L-TGF-ß, suggesting that tumor cell αvß8 serves as a platform for activating cell-surface L-TGF-ß presented by immune cells. Transcriptome analysis of tumor-associated lymphoid cells reveals macrophages as a key cell type responsive to ß8 inhibition with major increases in chemokine and tumor-eliminating genes. High ß8 expression in tumor cells is seen in 20%-80% of various cancers, which rarely coincides with high PD-L1 expression. These data suggest tumor cell αvß8 is a PD-1/PD-L1-independent immunotherapeutic target.


Subject(s)
Integrins/metabolism , Macrophages/immunology , Neoplasms/immunology , Transforming Growth Factor beta/metabolism , Tumor Escape/immunology , Animals , Antineoplastic Agents, Immunological/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Cell Line, Tumor , Computer Simulation , Disease Models, Animal , Female , Humans , Integrins/antagonists & inhibitors , Kaplan-Meier Estimate , Macrophages/metabolism , Male , Mice , Mice, Transgenic , Neoplasms/drug therapy , Neoplasms/mortality , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Tumor Escape/drug effects , Tumor Microenvironment/immunology
17.
Sci Transl Med ; 10(433)2018 03 21.
Article in English | MEDLINE | ID: mdl-29563320

ABSTRACT

Depending on age of acquisition, hepatitis B virus (HBV) can induce a cell-mediated immune response that results in either cure or progressive liver injury. In adult-acquired infection, HBV antigens are usually cleared, whereas in infancy-acquired infection, they persist. Individuals infected during infancy therefore represent the majority of patients chronically infected with HBV (CHB). A therapy that can promote viral antigen clearance in most CHB patients has not been developed and would represent a major health care advance and cost mitigator. Using an age-dependent mouse model of HBV clearance and persistence in conjunction with human blood and liver tissue, we studied mechanisms of viral clearance to identify new therapeutic targets. We demonstrate that age-dependent expression of the costimulatory molecule OX40 ligand (OX40L) by hepatic innate immune cells is pivotal in determining HBV immunity, and that treatment with OX40 agonists leads to improved HBV antigen clearance in young mice, as well as increased strength of T cell responses in young mice and adult mice that were exposed to HBV when they were young and developed a CHB serological profile. Similarly, in humans, we show that hepatic OX40L transcript expression is age-dependent and that increased OX40 expression on peripheral CD4+ T cells in adults is associated with HBV clearance. These findings provide new mechanistic understanding of the immune pathways and cells necessary for HBV immunity and identify potential therapeutic targets for resolving CHB.


Subject(s)
Hepatitis B virus/immunology , Hepatitis B virus/pathogenicity , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/metabolism , Immunity, Innate/physiology , OX40 Ligand/metabolism , Receptors, OX40/metabolism , Animals , Immunity, Innate/genetics , Mice , Mice, Knockout
18.
PLoS One ; 12(9): e0183912, 2017.
Article in English | MEDLINE | ID: mdl-28873429

ABSTRACT

Neutrophils and macrophages are important constituents of the hepatic inflammatory infiltrate in non-alcoholic steatohepatitis. These innate immune cells express CD18, an adhesion molecule that facilitates leukocyte activation. In the context of fatty liver, activation of infiltrated leukocytes is believed to enhance hepatocellular injury. The objective of this study was to determine the degree to which activated innate immune cells promote steatohepatitis by comparing hepatic outcomes in wild-type and CD18-mutant mice fed a methionine-choline-deficient (MCD) diet. After 3 weeks of MCD feeding, hepatocyte injury, based on serum ALT elevation, was 40% lower in CD18-mutant than wild-type mice. Leukocyte infiltration into the liver was not impaired in CD18-mutant mice, but leukocyte activation was markedly reduced, as shown by the lack of evidence of oxidant production. Despite having reduced hepatocellular injury, CD18-mutant mice developed significantly more hepatic steatosis than wild-type mice after MCD feeding. This coincided with greater hepatic induction of pro-inflammatory and lipogenic genes as well as a modest reduction in hepatic expression of adipose triglyceride lipase. Overall, the data indicate that CD18 deficiency curbs MCD-mediated liver injury by limiting the activation of innate immune cells in the liver without compromising intrahepatic cytokine activation. Reduced liver injury occurs at the expense of increased hepatic steatosis, which suggests that in addition to damaging hepatocytes, infiltrating leukocytes may influence lipid homeostasis in the liver.


Subject(s)
CD18 Antigens/genetics , CD18 Antigens/physiology , Fatty Liver/metabolism , Adipose Tissue/metabolism , Animals , Choline/chemistry , Cytokines/metabolism , Disease Models, Animal , Hepatocytes/cytology , Immunity, Innate , Inflammation , Leukocytes/cytology , Leukocytes/metabolism , Lipase/metabolism , Liver/metabolism , Male , Methionine/chemistry , Mice , Mice, Inbred C57BL , Mutation , Oxygen/chemistry , Peroxidase/metabolism , Triglycerides/metabolism
19.
Cell Mol Gastroenterol Hepatol ; 4(2): 223-236, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28649594

ABSTRACT

BACKGROUND & AIMS: The factors that distinguish metabolically healthy obesity from metabolically unhealthy obesity are not well understood. Diet has been implicated as a determinant of the unhealthy obesity phenotype, but which aspects of the diet induce dysmetabolism are unknown. The goal of this study was to investigate whether specific macronutrients or macronutrient combinations provoke dysmetabolism in the context of isocaloric, high-energy diets. METHODS: Mice were fed 4 high-energy diets identical in calorie and nutrient content but different in nutrient composition for 3 weeks to 6 months. The test diets contained 42% carbohydrate (sucrose or starch) and 42% fat (oleate or palmitate). Weight and glucose tolerance were monitored; blood and tissues were collected for histology, gene expression, and immunophenotyping. RESULTS: Mice gained weight on all 4 test diets but differed significantly in other metabolic outcomes. Animals fed the starch-oleate diet developed more severe hepatic steatosis than those on other formulas. Stable isotope incorporation showed that the excess hepatic steatosis in starch-oleate-fed mice derived from exaggerated adipose tissue lipolysis. In these mice, adipose tissue lipolysis coincided with adipocyte necrosis and inflammation. Notably, the liver and adipose tissue abnormalities provoked by starch-oleate feeding were reproduced when mice were fed a mixed-nutrient Western diet with 42% carbohydrate and 42% fat. CONCLUSIONS: The macronutrient composition of the diet exerts a significant influence on metabolic outcome, independent of calories and nutrient proportions. Starch-oleate appears to cause hepatic steatosis by inducing progressive adipose tissue injury. Starch-oleate phenocopies the effect of a Western diet; consequently, it may provide clues to the mechanism whereby specific nutrients cause metabolically unhealthy obesity.

20.
Sci Transl Med ; 6(241): 241ra79, 2014 Jun 18.
Article in English | MEDLINE | ID: mdl-24944194

ABSTRACT

Airway remodeling, caused by inflammation and fibrosis, is a major component of chronic obstructive pulmonary disease (COPD) and currently has no effective treatment. Transforming growth factor-ß (TGF-ß) has been widely implicated in the pathogenesis of airway remodeling in COPD. TGF-ß is expressed in a latent form that requires activation. The integrin αvß8 (encoded by the itgb8 gene) is a receptor for latent TGF-ß and is essential for its activation. Expression of integrin αvß8 is increased in airway fibroblasts in COPD and thus is an attractive therapeutic target for the treatment of airway remodeling in COPD. We demonstrate that an engineered optimized antibody to human αvß8 (B5) inhibited TGF-ß activation in transgenic mice expressing only human and not mouse ITGB8. The B5 engineered antibody blocked fibroinflammatory responses induced by tobacco smoke, cytokines, and allergens by inhibiting TGF-ß activation. To clarify the mechanism of action of B5, we used hydrodynamic, mutational, and electron microscopic methods to demonstrate that αvß8 predominantly adopts a constitutively active, extended-closed headpiece conformation. Epitope mapping and functional characterization of B5 revealed an allosteric mechanism of action due to locking-in of a low-affinity αvß8 conformation. Collectively, these data demonstrate a new model for integrin function and present a strategy to selectively target the TGF-ß pathway to treat fibroinflammatory airway diseases.


Subject(s)
Tracheitis/therapy , Transforming Growth Factor beta/metabolism , Animals , Humans , Mice , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL