Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
1.
Front Immunol ; 15: 1374828, 2024.
Article in English | MEDLINE | ID: mdl-39026668

ABSTRACT

Introduction: Innate lymphoid cells (ILCs) are enriched at mucosal surfaces where they respond rapidly to environmental stimuli and contribute to both tissue inflammation and healing. Methods: To gain insight into the role of ILCs in the pathology and recovery from COVID-19 infection, we employed a multi-omics approach consisting of Abseq and targeted mRNA sequencing to respectively probe the surface marker expression, transcriptional profile and heterogeneity of ILCs in peripheral blood of patients with COVID-19 compared with healthy controls. Results: We found that the frequency of ILC1 and ILC2 cells was significantly increased in COVID-19 patients. Moreover, all ILC subsets displayed a significantly higher frequency of CD69-expressing cells, indicating a heightened state of activation. ILC2s from COVID-19 patients had the highest number of significantly differentially expressed (DE) genes. The most notable genes DE in COVID-19 vs healthy participants included a) genes associated with responses to virus infections and b) genes that support ILC self-proliferation, activation and homeostasis. In addition, differential gene regulatory network analysis revealed ILC-specific regulons and their interactions driving the differential gene expression in each ILC. Discussion: Overall, this study provides mechanistic insights into the characteristics of ILC subsets activated during COVID-19 infection.


Subject(s)
COVID-19 , Immunity, Innate , Lymphocytes , Adult , Aged , Female , Humans , Male , Middle Aged , COVID-19/immunology , COVID-19/genetics , Gene Expression Profiling , Gene Regulatory Networks , Lymphocytes/immunology , Lymphocytes/metabolism , Multiomics , Single-Cell Analysis , Transcriptome
2.
Nat Commun ; 13(1): 6646, 2022 11 04.
Article in English | MEDLINE | ID: mdl-36333296

ABSTRACT

While food allergy oral immunotherapy (OIT) can provide safe and effective desensitization (DS), the immune mechanisms underlying development of sustained unresponsiveness (SU) following a period of avoidance are largely unknown. Here, we compare high dimensional phenotypes of innate and adaptive immune cell subsets of participants in a previously reported, phase 2 randomized, controlled, peanut OIT trial who achieved SU vs. DS (no vs. with allergic reactions upon food challenge after a withdrawal period; n = 21 vs. 30 respectively among total 120 intent-to-treat participants). Lower frequencies of naïve CD8+ T cells and terminally differentiated CD57+CD8+ T cell subsets at baseline (pre-OIT) are associated with SU. Frequency of naïve CD8+ T cells shows a significant positive correlation with peanut-specific and Ara h 2-specific IgE levels at baseline. Higher frequencies of IL-4+ and IFNγ+ CD4+ T cells post-OIT are negatively correlated with SU. Our findings provide evidence that an immune signature consisting of certain CD8+ T cell subset frequencies is potentially predictive of SU following OIT.


Subject(s)
Peanut Hypersensitivity , Peanut Hypersensitivity/therapy , Desensitization, Immunologic/methods , Immunoglobulin E , CD8-Positive T-Lymphocytes , Feasibility Studies , Administration, Oral , Arachis , Allergens , Immunologic Factors , Cell Differentiation
3.
Front Immunol ; 13: 960329, 2022.
Article in English | MEDLINE | ID: mdl-36420263

ABSTRACT

Graft-versus-host disease (GvHD) is a major complication after allogeneic hematopoietic cell transplantation (HCT). Current strategies to prevent GvHD with immunosuppressive drugs carry significant morbidity and may affect the graft-versus-tumor (GVT) effect. Inflammatory bowel disease (IBD) is an intestinal inflammatory condition that affects more than 2 million people in the United States. Current strategies to prevent colitis with immunosuppressive drugs carry significant morbidity. Recently, Repulsive Guidance Molecule b (RGMb) has been identified as part of a signaling hub with neogenin and BMP receptors in mice and humans. In addition, RGMb binds BMP-2/4 in mice and humans as well as PD-L2 in mice. RGMb is expressed in the gut epithelium and by antigen presenting cells, and we found significantly increased expression in mouse small intestine after total body irradiation HCT conditioning. We hypothesized that RGMb may play a role in GvHD and IBD pathogenesis by contributing to mucosal inflammation. Using major-mismatched HCT mouse models, treatment with an anti-RGMb monoclonal antibody (mAb) that blocks the interaction with BMP-2/4 and neogenin prevented GvHD and improved survival compared to isotype control (75% versus 30% survival at 60 days after transplantation). The GVT effect was retained in tumor models. Using an inflammatory bowel disease dextran sulfate sodium model, treatment with anti-RGMb blocking monoclonal antibody but not isotype control prevented colitis and improved survival compared to control (73% versus 33% at 21 days after treatment) restoring gut homeostasis. Anti-RGMb mAb (9D1) treatment decreased IFN-γ and significantly increased IL-5 and IL-10 in the gut of the treated mice compared to the isotype control treated mice.


Subject(s)
Colitis , Graft vs Host Disease , Inflammatory Bowel Diseases , Humans , Mice , Animals , Inflammation , Inflammatory Bowel Diseases/therapy , Colitis/chemically induced , Immunosuppressive Agents , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Cell Adhesion Molecules, Neuronal
4.
J Clin Invest ; 132(20)2022 10 17.
Article in English | MEDLINE | ID: mdl-36250466

ABSTRACT

Food allergies are a leading cause of anaphylaxis, and allergen-specific immune responses in both the innate and the adaptive immune system play key roles in its pathogenesis. We conducted a comprehensive phenotypic and functional investigation of immune cell responses from nonallergic (NA) and peanut allergic (PA) participants cultured with media alone or peanut protein and found, surprisingly, that NK cell activation was strongly associated with the immune response to allergen in PA participants. Peanut-responsive NK cells manifested a distinct expression pattern in PA participants compared with NA participants. Allergen-activated NK cells expressed both Th2 and immune regulatory cytokines, hinting at a potential functional role in mediating and regulating the Th2 allergic response. Depletion of CD3+ T cells attenuated the response of NK cells to peanut-allergen stimulation, suggesting that peanut-responsive NK cells are T cell dependent. We also showed that oral immune therapy was associated with decreased NK responses to peanut allergen stimulation in vitro. These results demonstrate that NK cells are associated with the food-allergic immune response, and the magnitude of this mobilized cell population suggests that they play a functional role in allergic immunity.


Subject(s)
Food Hypersensitivity , Peanut Hypersensitivity , Allergens , Arachis , Cytokines/metabolism , Humans , Killer Cells, Natural , Peanut Hypersensitivity/therapy
6.
Allergy ; 76(9): 2809-2826, 2021 09.
Article in English | MEDLINE | ID: mdl-33782956

ABSTRACT

BACKGROUND: Multifood oral immunotherapy (mOIT) with adjunctive anti-IgE (omalizumab, XOLAIR® ) treatment affords safe, effective, and rapid desensitization to multiple foods, although the specific immune mechanisms mediating this desensitization remain to be fully elucidated. METHODS: Participants in our phase 2 mOIT trial (NCT02643862) received omalizumab from baseline to week 16 and mOIT from week 8 to week 36. We compared the immune profile of PBMCs and plasma taken at baseline, week 8, and week 36 using high-dimensional mass cytometry, component-resolved diagnostics, the indirect basophil activation test, and Luminex. RESULTS: We found (i) decreased frequency of IL-4+ peanut-reactive CD4+ T cells and a marked downregulation of GPR15 expression and CXCR3 frequency among γδ and CD8+ T-cell subsets at week 8 during the initial, omalizumab-alone induction phase; (ii) significant upregulation of the skin-homing receptor CCR4 in peanut-reactive CD4+ T and Th2 effector memory (EM) cells and of cutaneous lymphocyte-associated antigen (CLA) in peanut-reactive CD8+ T and CD8+ EM cells; (iii) downregulation of CD86 expression among antigen-presenting cell subsets; and (iv) reduction in pro-inflammatory cytokines, notably IL-17, at week 36 post-OIT. We also observed significant attenuation of the Th2 phenotype post-OIT, defined by downregulation of IL-4 peanut-reactive T cells and OX40 in Th2EM cells, increased allergen component-specific IgG4/IgE ratio, and decreased allergen-driven activation of indirectly sensitized basophils. CONCLUSIONS: This exploratory study provides novel comprehensive insight into the immune underpinnings of desensitization through omalizumab-facilitated mOIT. Moreover, this study provides encouraging results to support the complex immune changes that can be induced by OIT.


Subject(s)
Omalizumab , Peanut Hypersensitivity , Administration, Oral , Allergens , Desensitization, Immunologic , Humans , Immunoglobulin E , Omalizumab/therapeutic use
7.
Adv Exp Med Biol ; 1253: 153-181, 2020.
Article in English | MEDLINE | ID: mdl-32445095

ABSTRACT

Asthma and rhinitis are complex, heterogeneous diseases characterized by chronic inflammation of the upper and lower airways. While genome-wide association studies (GWAS) have identified a number of susceptible loci and candidate genes associated with the pathogenesis of asthma and allergic rhinitis (AR), the risk-associated alleles account for only a very small percent of the genetic risk. In allergic airway and other complex diseases, it is thought that epigenetic modifications, including DNA methylation, histone modifications, and non-coding microRNAs, caused by complex interactions between the underlying genome and the environment may account for some of this "missing heritability" and may explain the high degree of plasticity in immune responses. In this chapter, we will focus on the current knowledge of classical epigenetic modifications, DNA methylation and histone modifications, and their potential role in asthma and AR. In particular, we will review epigenetic variations associated with maternal airway disease, demographics, environment, and non-specific associations. The role of specific genetic haplotypes in environmentally induced epigenetic changes are also discussed. A major limitation of many of the current studies of asthma epigenetics is that they evaluate epigenetic modifications in both allergic and non-allergic asthma, making it difficult to distinguish those epigenetic modifications that mediate allergic asthma from those that mediate non-allergic asthma. Additionally, most DNA methylation studies in asthma use peripheral or cord blood due to poor accessibility of airway cells or tissue. Unlike DNA sequences, epigenetic alterations are quite cell- and tissue-specific, and epigenetic changes found in airway tissue or cells may be discordant from that of circulating blood. These two confounding factors should be considered when reviewing epigenetic studies in allergic airway disease.


Subject(s)
Asthma/genetics , Epigenesis, Genetic , Epigenomics , Gene-Environment Interaction , Rhinitis, Allergic/genetics , Genome-Wide Association Study , Humans
9.
J Allergy Clin Immunol ; 145(4): 1072-1081.e1, 2020 04.
Article in English | MEDLINE | ID: mdl-31926182

ABSTRACT

The aims of the Keystone Symposium conference, "Origins of allergic disease: Microbial, epithelial and immune interactions" were to present and discuss potential microbial-epithelial-immune interactions underlying the early-life origins of allergic disorders, as well as immune mechanisms that might suggest novel disease prevention or intervention strategies. Cross-talk and sharing of ideas among participating experts in basic science and clinical aspects of allergic diseases provided substantial insight into the concept of allergic disorders as a systems disease. The overriding message distilled from the discussions was that damage to epithelial surfaces lies at the origin of the various manifestations of allergic disease. The epithelium of the lungs, gut, and skin, which operates as a critical sensor of environmental stimuli, is besieged by an onslaught of contemporary environmental forces including an altered microbiome, air pollution, food allergens in a changed diet, and chemicals in modern detergents. Collectively, this onslaught leads to alterations of lung, skin, or gut epithelial surfaces, driving a type 2 immune response that underlies most, if not all, of the atopic diseases. Possible remedies for treatment and prevention of allergic diseases were discussed, including a precision medicine approach using biologics, oral desensitization, targeted gut microbiome alterations, and behavior alteration.


Subject(s)
Asthma/immunology , Desensitization, Immunologic/methods , Epithelium/immunology , Hypersensitivity/immunology , Microbiota/immunology , Allergens/immunology , Animals , Asthma/metabolism , Host-Parasite Interactions , Humans , Hypersensitivity/microbiology , United States
10.
Blood Adv ; 3(21): 3419-3431, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31714958

ABSTRACT

Acute graft-versus-host disease (GVHD) is a leading cause of mortality after allogeneic hematopoietic cell transplantation (HCT) mediated by dysregulated T-cell immune reconstitution. Given the role of the T-cell immunoglobulin and mucin 1 (TIM-1) surface protein in many immune processes, including organ transplantation tolerance, we asked if TIM-1 might drive post-transplant inflammation and acute GVHD. TIM-1 binds to phosphatidylserine (PtdSer), and agonism of TIM1 on immune cells is proinflammatory. HCT conditioning results in a significant supply of PtdSer from apoptosis and cellular debris. Using murine models, treatment with an antagonistic anti-TIM-1 monoclonal antibody (mAb) protects against acute GVHD while maintaining graft-versus-tumor effects. In contrast, the addition of exogenous free PtdSer worsened GVHD in a TIM-1-dependent manner. Importantly, TIM-1 blockade did not alter the expansion of donor T cells in vitro or in vivo. Instead, TIM-1 blockade reduces proinflammatory cytokines and promotes anti-inflammatory factors like carbonic anhydrase 1 and serum amyloid A1 in the gut tissue. This is mediated by TIM-1 on donor cells, as HCT of wild-type (WT) bone marrow (BM) and conventional T (Tcon) cells into TIM-1-/- knockout (KO) recipient mice showed little survival advantage compared with WT recipients, whereas WT recipients of TIM-1-/- KO Tcon cells or TIM1-/- KO BM had improved survival, in part due to the expression of TIM-1 on donor invariant natural killer T cells, which drives inflammation. Finally, in a humanized mouse xenograft GVHD model, treatment with anti-human TIM-1 antagonist mAb reduced GVHD disease burden and mortality. This supports TIM-1 as important for GVHD pathogenesis and as a target for the prevention of GVHD.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Graft vs Host Disease/drug therapy , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Hepatitis A Virus Cellular Receptor 1/antagonists & inhibitors , Animals , Antibodies, Blocking/therapeutic use , Biomarkers , Disease Models, Animal , Gene Expression , Graft vs Host Disease/diagnosis , Graft vs Host Disease/mortality , Hematopoietic Stem Cell Transplantation/methods , Hepatitis A Virus Cellular Receptor 1/genetics , Hepatitis A Virus Cellular Receptor 1/metabolism , Humans , Immune Reconstitution , Immunohistochemistry , Immunophenotyping , Inflammation Mediators/metabolism , Lymphocyte Count , Mice , Mice, Knockout , Severity of Illness Index , Survival Rate , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transplantation, Homologous
11.
J Allergy Clin Immunol ; 144(1): 94-108.e11, 2019 07.
Article in English | MEDLINE | ID: mdl-30703386

ABSTRACT

BACKGROUND: Allergic asthma causes morbidity in many subjects, and novel precision-directed treatments would be valuable. OBJECTIVE: We sought to examine the role of a novel innate molecule, repulsive guidance molecule b (RGMb), in murine models of allergic asthma. METHODS: In models of allergic asthma using ovalbumin or cockroach allergen, mice were treated with anti-RGMb or control mAb and examined for airway inflammation and airway hyperreactivity (AHR), a cardinal feature of asthma. The mechanisms by which RGMb causes airways disease were also examined. RESULTS: We found that blockade of RGMb by treatment with anti-RGMb mAb effectively blocked the development of airway inflammation and AHR. Importantly, blockade of RGMb completely blocked the development of airway inflammation and AHR, even if treatment occurred only during the challenge (effector) phase. IL-25 played an important role in these models of asthma because IL-25 receptor-deficient mice did not develop disease after sensitization and challenge with allergen. RGMb was expressed primarily by innate cells in the lungs, including bronchial epithelial cells (known producers of IL-25), activated eosinophils, and interstitial macrophages, which in the inflamed lung expressed the IL-25 receptor and produced IL-5 and IL-13. We also found that neogenin, the canonical receptor for RGMb, was expressed by interstitial macrophages and bronchial epithelial cells in the inflamed lung, suggesting that an innate RGMb-neogenin axis might modulate allergic asthma. CONCLUSIONS: These results demonstrate an important role for a novel innate pathway in regulating type 2 inflammation in patients with allergic asthma involving RGMb and RGMb-expressing cells, such as interstitial macrophages and bronchial epithelial cells. Moreover, targeting this previously unappreciated innate pathway might provide an important treatment option for allergic asthma.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Asthma/drug therapy , Bronchial Hyperreactivity/drug therapy , Cell Adhesion Molecules, Neuronal/antagonists & inhibitors , Allergens/immunology , Animals , Asthma/immunology , Bronchial Hyperreactivity/immunology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cell Adhesion Molecules, Neuronal/immunology , Cockroaches/immunology , Female , Interleukin-1 Receptor-Like 1 Protein/genetics , Macrophages/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/immunology , Programmed Cell Death 1 Ligand 2 Protein/genetics , Receptors, Interleukin/genetics , Receptors, Interleukin/immunology
12.
J Allergy Clin Immunol ; 143(2): 565-576.e7, 2019 02.
Article in English | MEDLINE | ID: mdl-29852257

ABSTRACT

BACKGROUND: Infection of suckling mice with influenza virus expands a CD4-CD8- double-negative (DN) natural killer T (NKT) cell subpopulation that protects the mice as adults against allergen-induced airway hyperreactivity (AHR). However, this NKT cell subset has not been characterized, and the underlying mechanisms of protection remain unknown. OBJECTIVE: We characterized this specific NKT cell subpopulation that developed during influenza infection in neonatal mice and that suppressed the subsequent development of AHR. METHODS: A cell-surface marker was identified by comparing the mRNA expression profile of wild-type CD4+ NKT cells with that of suppressive Vα14 DN NKT cells. The marker-enriched NKT cell subset was then analyzed for its cytokine profile and its suppressive in vitro and in vivo abilities. RESULTS: We showed that DN NKT cells with high CD38 expression produced IFN-γ, but not IL-17, IL-4, or IL-13, and inhibited development of AHR through contact-dependent suppression of helper CD4 T-cell proliferation. The NKT subset expanded in the lungs of neonatal mice after infection with influenza and also after treatment of neonatal mice with Nu-α-GalCer, which effectively increased DN CD38hi NKT cell numbers. CONCLUSION: These results suggest that early/neonatal exposure to infection or antigen challenge affects subsequent lung immunity by altering the cellular composition of cells in the lung and that some subsets of NKT cells suppress AHR. These results provide a possible mechanism by which prior infections can protect against the development of allergic asthma and might be further explored as a protective measure for young children.


Subject(s)
Asthma/immunology , Influenza A virus/physiology , Influenza, Human/immunology , Lung/immunology , Natural Killer T-Cells/immunology , Orthomyxoviridae Infections/immunology , Respiratory Hypersensitivity/immunology , Animals , Animals, Newborn , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Disease Resistance , Humans , Immune Tolerance , Immunity, Maternally-Acquired , Lung/virology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Transcriptome
13.
Pediatr Allergy Immunol ; 28(7): 619-627, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28782296

ABSTRACT

Food allergy is a significant medical problem that affects up to 8% of children in developed countries. At present, there are no curative therapies available in routine practice and management of food allergy involves strict allergen avoidance, education, and prompt treatment upon accidental exposure. Oral immunotherapy (OIT) is an efficacious experimental approach to food allergy and has been shown to provide a substantial benefit in terms of allergen desensitization. However, OIT is associated with high rates of allergic reactions, and the period of protection offered by OIT appears to be limited and highly variable. Recurrence of allergen sensitivity after a period of treatment discontinuation is commonly observed. With the aim of overcoming these limitations of OIT, several trials have studied omalizumab (anti-IgE monoclonal antibody) as an adjuvant treatment for patients undergoing OIT. Results from these trials have shown that the addition of omalizumab to OIT leads to a significant decrease in the frequency and severity of reactions, which allows for an increase in the threshold of tolerance to food allergens. This review provides a summary of the current literature and addresses some of the key questions that remain regarding the use of omalizumab in conjunction with OIT.


Subject(s)
Anti-Allergic Agents/therapeutic use , Antibodies, Anti-Idiotypic/immunology , Food Hypersensitivity/drug therapy , Immunotherapy/adverse effects , Omalizumab/therapeutic use , Administration, Oral , Adolescent , Child , Desensitization, Immunologic/methods , Drug Therapy, Combination , Food Hypersensitivity/immunology , Humans , Immune Tolerance , Immunotherapy/methods , Young Adult
14.
Sci Rep ; 7(1): 1878, 2017 05 12.
Article in English | MEDLINE | ID: mdl-28500324

ABSTRACT

Extracellular vesicles (EVs), including exosomes and microvesicles, are 30-800 nm vesicles that are released by most cell types, as biological packages for intercellular communication. Their importance in cancer and inflammation makes EVs and their cargo promising biomarkers of disease and cell-free therapeutic agents. Emerging high-resolution cytometric methods have created a pressing need for efficient fluorescent labeling procedures to visualize and detect EVs. Suitable labels must be bright enough for one EV to be detected without the generation of label-associated artifacts. To identify a strategy that robustly labels individual EVs, we used nanoFACS, a high-resolution flow cytometric method that utilizes light scattering and fluorescence parameters along with sample enumeration, to evaluate various labels. Specifically, we compared lipid-, protein-, and RNA-based staining methods and developed a robust EV staining strategy, with the amine-reactive fluorescent label, 5-(and-6)-Carboxyfluorescein Diacetate Succinimidyl Ester, and size exclusion chromatography to remove unconjugated label. By combining nanoFACS measurements of light scattering and fluorescence, we evaluated the sensitivity and specificity of EV labeling assays in a manner that has not been described for other EV detection methods. Efficient characterization of EVs by nanoFACS paves the way towards further study of EVs and their roles in health and disease.


Subject(s)
Extracellular Vesicles/metabolism , Flow Cytometry , Staining and Labeling , Amines , Animals , Cell Line , Cell-Derived Microparticles/metabolism , Chromatography, Gel , Flow Cytometry/methods , Fluorescent Dyes , Mice , Micelles , Staining and Labeling/methods
15.
Curr Allergy Asthma Rep ; 16(12): 87, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27943047

ABSTRACT

PURPOSE OF REVIEW: The prevalence and severity of IgE-mediated food allergy has increased dramatically over the last 15 years and is becoming a global health problem. Multiple lines of evidence suggest that epigenetic modifications of the genome resulting from gene-environment interactions have a key role in the increased prevalence of atopic disease. In this review, we describe the recent evidence suggesting how epigenetic changes mediate susceptibility to food allergies, and discuss how immunotherapy (IT) may reverse these effects. We discuss the areas of the epigenome as yet unexplored in terms of food allergy and IT such as histone modification and chromatin accessibility, and new techniques that may be utilized in future studies. RECENT FINDINGS: Recent findings provide strong evidence that DNA methylation of certain promoter regions such as Forkhead box protein 3 is associated with clinical reactivity, and further, can be changed during IT treatment. Reports on other epigenetic changes are limited but also show evidence of significant change based on both disease status and treatment. In comparison to epigenetic studies focusing on asthma and allergic rhinitis, food allergy remains understudied. However, within the next decade, it is likely that epigenetic modifications may be used as biomarkers to aid in diagnosis and treatment of food-allergic patients. DNA methylation at specific loci has shown associations between food challenge outcomes, successful desensitization treatment, and overall phenotype compared to healthy controls.


Subject(s)
Food Hypersensitivity/genetics , Food Hypersensitivity/therapy , Immunotherapy/methods , DNA Methylation , Epigenesis, Genetic , Food Hypersensitivity/immunology , Humans
16.
Eur J Immunol ; 46(4): 795-806, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26891006

ABSTRACT

Asthma is a complex and heterogeneous disease that is characterized by airway hyper-reactivity (AHR) and airway inflammation. Although asthma was long thought to be driven by allergen-reactive TH 2 cells, it has recently become clear that the pathogenesis of asthma is more complicated and associated with multiple pathways and cell types. A very exciting recent development was the discovery of innate lymphoid cells (ILCs) as key players in the pathogenesis of asthma. ILCs do not express antigen receptors but react promptly to "danger signals" from inflamed tissue and produce an array of cytokines that direct the ensuing immune response. The roles of ILCs may differ in distinct asthma phenotypes. ILC2s may be critical for initiation of adaptive immune responses in inhaled allergen-driven AHR, but may also function independently of adaptive immunity, mediating influenza-induced AHR. ILC2s also contribute to resolution of lung inflammation through their production of amphiregulin. Obesity-induced asthma is associated with expansion of IL-17A-producing ILC3s in the lungs. Furthermore, ILCs may also contribute to steroid-resistant asthma. Although the precise roles of ILCs in different types of asthma are still under investigation, it is clear that inhibition of ILC function represents a potential target that could provide novel treatments for asthma.


Subject(s)
Adaptive Immunity/immunology , Asthma/immunology , Immunity, Innate/immunology , Lymphocytes/immunology , Allergens/immunology , Amphiregulin , Asthma/pathology , EGF Family of Proteins/immunology , Humans , Influenza, Human/immunology , Interleukin-17/immunology , Interleukin-33/immunology , Lung/cytology , Lung/immunology , Lung/pathology , Obesity/immunology , Signal Transduction/immunology
17.
Arterioscler Thromb Vasc Biol ; 36(3): 456-65, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26821944

ABSTRACT

OBJECTIVE: T cell immunoglobulin and mucin domain (Tim) proteins are expressed by numerous immune cells, recognize phosphatidylserine on apoptotic cells, and function as costimulators or coinhibitors. Tim-1 is expressed by activated T cells but is also found on dendritic cells and B cells. Tim-4, present on macrophages and dendritic cells, plays a critical role in apoptotic cell clearance, regulates the number of phosphatidylserine-expressing activated T cells, and is genetically associated with low low-density lipoprotein and triglyceride levels. Because these functions of Tim-1 and Tim-4 could affect atherosclerosis, their modulation has potential therapeutic value in cardiovascular disease. APPROACH AND RESULTS: ldlr(-/-) mice were fed a high-fat diet for 4 weeks while being treated with control (rat immunoglobulin G1) or anti-Tim-1 (3D10) or -Tim-4 (21H12) monoclonal antibodies that block phosphatidylserine recognition and phagocytosis. Both anti-Tim-1 and anti-Tim-4 treatments enhanced atherosclerosis by 45% compared with controls by impairment of efferocytosis and increasing aortic CD4(+)T cells. Consistently, anti-Tim-4-treated mice showed increased percentages of activated T cells and late apoptotic cells in the circulation. Moreover, in vitro blockade of Tim-4 inhibited efferocytosis of oxidized low-density lipoprotein-induced apoptotic macrophages. Although anti-Tim-4 treatment increased T helper cell (Th)1 and Th2 responses, anti-Tim-1 induced Th2 responses but dramatically reduced the percentage of regulatory T cells. Finally, combined blockade of Tim-1 and Tim-4 increased atherosclerotic lesion size by 59%. CONCLUSIONS: Blockade of Tim-4 aggravates atherosclerosis likely by prevention of phagocytosis of phosphatidylserine-expressing apoptotic cells and activated T cells by Tim-4-expressing cells, whereas Tim-1-associated effects on atherosclerosis are related to changes in Th1/Th2 balance and reduced circulating regulatory T cells.


Subject(s)
Antibodies, Monoclonal/toxicity , Aortic Diseases/chemically induced , Atherosclerosis/chemically induced , CD4-Positive T-Lymphocytes/drug effects , Macrophages/drug effects , Membrane Proteins/antagonists & inhibitors , Receptors, LDL/deficiency , Animals , Aortic Diseases/genetics , Aortic Diseases/immunology , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apoptosis/drug effects , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Cell Proliferation/drug effects , Cells, Cultured , Coculture Techniques , Diet, High-Fat , Disease Models, Animal , Female , Hepatitis A Virus Cellular Receptor 1 , Lipoproteins, LDL/metabolism , Lymphocyte Activation/drug effects , Macrophages/immunology , Macrophages/pathology , Male , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice, Knockout , Phagocytosis/drug effects , Plaque, Atherosclerotic , Receptors, LDL/genetics , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Th1 Cells/drug effects , Th1 Cells/immunology , Th2 Cells/drug effects , Th2 Cells/immunology
18.
Traffic ; 16(11): 1193-207, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26332704

ABSTRACT

The T cell costimulatory molecule TIM-1 (T cell/transmembrane, mucin and immunoglobulin domain protein 1) sorts mainly to endosomes in lymphoid cells. At difference from the cell surface protein, endosomal TIM-1 translocates to the immune synapse (IS), where it can contribute to antigen-dependent T cell costimulation. TIM-1 ligands increase the amount of cell surface protein, preventing its traffic to the IS. The bipolar sorting of TIM-1 observed during IS formation is determined by differences in its subcellular location, and probably modulates antigen-driven immune responses.


Subject(s)
Cell Membrane/metabolism , Cell Movement/physiology , Endosomes/metabolism , Membrane Glycoproteins/metabolism , Receptors, Virus/metabolism , Synapses/metabolism , Hepatitis A Virus Cellular Receptor 1 , Humans , Protein Transport/physiology , Signal Transduction/immunology , Synapses/immunology , T-Lymphocytes/immunology
19.
Immunol Rev ; 260(1): 235-48, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24942693

ABSTRACT

The lung, while functioning as a gas exchange organ, encounters a large array of environmental factors, including particulate matter, toxins, reactive oxygen species, chemicals, allergens, and infectious microbes. To rapidly respond to and counteract these elements, a number of innate immune mechanisms have evolved that can lead to lung inflammation and asthma, which is the focus of this review. These innate mechanisms include a role for two incompletely understood cell types, invariant natural killer T (iNKT) cells and innate lymphoid cells (ILCs), which together produce a wide range of cytokines, including interleukin-4 (IL-4), IL-5, IL-13, interferon-γ, IL-17, and IL-22, independently of adaptive immunity and conventional antigens. The specific roles of iNKT cells and ILCs in immunity are still being defined, but both cell types appear to play important roles in the lungs, particularly in asthma. As we gain a better understanding of these innate cell types, we will acquire great insight into the mechanisms by which allergic and non-allergic asthma phenotypes develop.


Subject(s)
Asthma/immunology , Immunity, Innate , Lung/immunology , Adaptive Immunity , Allergens/immunology , Animals , Asthma/genetics , Asthma/metabolism , Asthma/microbiology , Cell Communication , Humans , Lung/metabolism , Lung/microbiology , Lymphocytes/immunology , Lymphocytes/metabolism , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Respiratory Hypersensitivity/genetics , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/metabolism , Respiratory Hypersensitivity/microbiology
20.
J Exp Med ; 211(5): 943-59, 2014 May 05.
Article in English | MEDLINE | ID: mdl-24752301

ABSTRACT

We report that programmed death ligand 2 (PD-L2), a known ligand of PD-1, also binds to repulsive guidance molecule b (RGMb), which was originally identified in the nervous system as a co-receptor for bone morphogenetic proteins (BMPs). PD-L2 and BMP-2/4 bind to distinct sites on RGMb. Normal resting lung interstitial macrophages and alveolar epithelial cells express high levels of RGMb mRNA, whereas lung dendritic cells express PD-L2. Blockade of the RGMb-PD-L2 interaction markedly impaired the development of respiratory tolerance by interfering with the initial T cell expansion required for respiratory tolerance. Experiments with PD-L2-deficient mice showed that PD-L2 expression on non-T cells was critical for respiratory tolerance, but expression on T cells was not required. Because PD-L2 binds to both PD-1, which inhibits antitumor immunity, and to RGMb, which regulates respiratory immunity, targeting the PD-L2 pathway has therapeutic potential for asthma, cancer, and other immune-mediated disorders. Understanding this pathway may provide insights into how to optimally modulate the PD-1 pathway in cancer immunotherapy while minimizing adverse events.


Subject(s)
Immune Tolerance/immunology , Lung/immunology , Nerve Tissue Proteins/metabolism , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Animals , Bone Morphogenetic Protein 2/metabolism , Cell Adhesion Molecules, Neuronal , Cell Line, Tumor , Epithelial Cells/metabolism , GPI-Linked Proteins , Humans , Lung/metabolism , Macrophages, Alveolar/metabolism , Mice , Nerve Tissue Proteins/immunology , Programmed Cell Death 1 Ligand 2 Protein/immunology , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...