Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Nat Immunol ; 20(9): 1138-1149, 2019 09.
Article in English | MEDLINE | ID: mdl-31427775

ABSTRACT

Interleukin (IL)-1R3 is the co-receptor in three signaling pathways that involve six cytokines of the IL-1 family (IL-1α, IL-1ß, IL-33, IL-36α, IL-36ß and IL-36γ). In many diseases, multiple cytokines contribute to disease pathogenesis. For example, in asthma, both IL-33 and IL-1 are of major importance, as are IL-36 and IL-1 in psoriasis. We developed a blocking monoclonal antibody (mAb) to human IL-1R3 (MAB-hR3) and demonstrate here that this antibody specifically inhibits signaling via IL-1, IL-33 and IL-36 in vitro. Also, in three distinct in vivo models of disease (crystal-induced peritonitis, allergic airway inflammation and psoriasis), we found that targeting IL-1R3 with a single mAb to mouse IL-1R3 (MAB-mR3) significantly attenuated heterogeneous cytokine-driven inflammation and disease severity. We conclude that in diseases driven by multiple cytokines, a single antagonistic agent such as a mAb to IL-1R3 is a therapeutic option with considerable translational benefit.


Subject(s)
Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , Interleukin-1 Receptor Accessory Protein/antagonists & inhibitors , Peritonitis/immunology , Pneumonia/immunology , Psoriasis/immunology , A549 Cells , Animals , Cell Line, Tumor , Disease Models, Animal , HEK293 Cells , Humans , Imiquimod/toxicity , Inflammation/pathology , Interleukin-1/immunology , Interleukin-1 Receptor Accessory Protein/immunology , Interleukin-1beta/immunology , Interleukin-33/immunology , Male , Mice , Mice, Inbred C57BL , Ovalbumin/toxicity , Peritonitis/drug therapy , Peritonitis/pathology , Pneumonia/drug therapy , Pneumonia/pathology , Psoriasis/drug therapy , Psoriasis/pathology , Signal Transduction/immunology , Uric Acid/toxicity
2.
J Immunol ; 208(8): 1835-1843, 2022 04 15.
Article in English | MEDLINE | ID: mdl-35418504

ABSTRACT

Sarcoidosis and chronic beryllium disease are noninfectious lung diseases that are characterized by the presence of noncaseating granulomatous inflammation. Chronic beryllium disease is caused by occupational exposure to beryllium containing particles, whereas the etiology of sarcoidosis is not known. Genetic susceptibility for both diseases is associated with particular MHC class II alleles, and CD4+ T cells are implicated in their pathogenesis. The innate immune system plays a critical role in the initiation of pathogenic CD4+ T cell responses as well as the transition to active lung disease and disease progression. In this review, we highlight recent insights into Ag recognition in chronic beryllium disease and sarcoidosis. In addition, we discuss the current understanding of the dynamic interactions between the innate and adaptive immune systems and their impact on disease pathogenesis.


Subject(s)
Berylliosis , Lung Diseases , Sarcoidosis , Adaptive Immunity , Beryllium , Chronic Disease , Granuloma , Humans , Sarcoidosis/complications
3.
Am J Respir Cell Mol Biol ; 64(5): 547-556, 2021 05.
Article in English | MEDLINE | ID: mdl-33332993

ABSTRACT

Over the last several decades, our understanding of regulated-cell-death (RCD) pathways has increased dramatically. In addition to apoptosis and accidental cell death (primary necrosis), a diverse spectrum of RCD pathways has been delineated. In the lung, airway macrophages are critical for maintaining the functionality of airways via the clearance of inhaled particles, cell debris, and infectious agents. Exposure of these cells to pathogenic organisms or particles can induce a variety of RCD pathways that promote the release of danger signals into the lung. These responses have evolved to trigger the innate and adaptive arms of the immune system and thus offer protection against pathogens; yet they can also contribute to the development of lung injury and pathogenic immune responses. In this review, we discuss recent studies that suggest a critical role for airway-macrophage RCD pathways in promoting the release of pulmonary danger signals in health and disease.


Subject(s)
Alarmins/immunology , Apoptosis Regulatory Proteins/immunology , Lung Injury/immunology , Lung/immunology , Macrophages, Alveolar/immunology , Respiratory Distress Syndrome/immunology , Adaptive Immunity , Alarmins/genetics , Animals , Apoptosis/genetics , Apoptosis/immunology , Apoptosis Regulatory Proteins/genetics , Cytokines/genetics , Cytokines/immunology , Epithelial Cells/immunology , Epithelial Cells/pathology , Gene Expression Regulation , Humans , Immunity, Innate , Lung/pathology , Lung Injury/genetics , Lung Injury/pathology , Macrophages, Alveolar/pathology , Necrosis/genetics , Necrosis/immunology , Necrosis/pathology , Pyroptosis/genetics , Pyroptosis/immunology , Respiratory Distress Syndrome/genetics , Respiratory Distress Syndrome/pathology , Signal Transduction
4.
J Immunol ; 201(8): 2232-2243, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30185516

ABSTRACT

Metal-induced hypersensitivity is driven by dendritic cells (DCs) that migrate from the site of exposure to the lymph nodes, upregulate costimulatory molecules, and initiate metal-specific CD4+ T cell responses. Chronic beryllium disease (CBD), a life-threatening metal-induced hypersensitivity, is driven by beryllium-specific CD4+ Th1 cells that expand in the lung-draining lymph nodes (LDLNs) after beryllium exposure (sensitization phase) and are recruited back to the lung, where they orchestrate granulomatous lung disease (elicitation phase). To understand more about how beryllium exposures impact DC function during sensitization, we examined the early events in the lung and LDLNs after pulmonary exposure to different physiochemical forms of beryllium. Exposure to soluble or crystalline forms of beryllium induced alveolar macrophage death/release of IL-1α and DNA, enhanced migration of CD80hi DCs to the LDLNs, and sensitized HLA-DP2 transgenic mice after single low-dose exposures, whereas exposures to insoluble particulate forms beryllium did not. IL-1α and DNA released by alveolar macrophages upregulated CD80 on immature BMDC via IL-1R1 and TLR9, respectively. Intrapulmonary exposure of mice to IL-1R and TLR9 agonists without beryllium was sufficient to drive accumulation of CD80hi DCs in the LDLNs, whereas blocking both pathways prevented accumulation of CD80hi DCs in the LDLNs of beryllium-exposed mice. Thus, in contrast to particulate forms of beryllium, which are poor sensitizers, soluble or crystalline forms of beryllium promote death of alveolar macrophages and their release of IL-1α and DNA, which act as damage-associated molecular pattern molecules to enhance DC function during beryllium sensitization.


Subject(s)
Berylliosis/immunology , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Hypersensitivity/immunology , Lung/pathology , Receptors, Interleukin-1 Type I/metabolism , Toll-Like Receptor 9/metabolism , Allergens/immunology , Animals , Beryllium/immunology , Cell Differentiation , Cell Movement , Cells, Cultured , Chronic Disease , Enzyme-Linked Immunospot Assay , Humans , Immunization , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics
5.
J Immunol ; 196(1): 22-7, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26685315

ABSTRACT

Chronic beryllium (Be) disease is a granulomatous lung disorder that results from Be exposure in a genetically susceptible host. The disease is characterized by the accumulation of Be-responsive CD4(+) T cells in the lung, and genetic susceptibility is primarily linked to HLA-DPB1 alleles possessing a glutamic acid at position 69 of the ß-chain. Recent structural analysis of a Be-specific TCR interacting with a Be-loaded HLA-DP2-peptide complex revealed that Be is coordinated by amino acid residues derived from the HLA-DP2 ß-chain and peptide and showed that the TCR does not directly interact with the Be(2+) cation. Rather, the TCR recognizes a modified HLA-DP2-peptide complex with charge and conformational changes. Collectively, these findings provide a structural basis for the development of this occupational lung disease through the ability of Be to induce posttranslational modifications in preexisting HLA-DP2-peptide complexes, resulting in the creation of neoantigens.


Subject(s)
Berylliosis/genetics , Berylliosis/immunology , Beryllium/immunology , CD4-Positive T-Lymphocytes/immunology , HLA-DP beta-Chains/immunology , Genetic Predisposition to Disease , HLA-DP beta-Chains/genetics , Humans , Hypersensitivity/genetics , Hypersensitivity/immunology , Lung/immunology , Lung/pathology , Protein Processing, Post-Translational/genetics , Receptors, Antigen, T-Cell/immunology
6.
Proc Natl Acad Sci U S A ; 111(23): 8553-8, 2014 Jun 10.
Article in English | MEDLINE | ID: mdl-24912188

ABSTRACT

Susceptibility to chronic beryllium disease (CBD) is linked to certain HLA-DP molecules, including HLA-DP2. To elucidate the molecular basis of this association, we exposed mice transgenic (Tg) for HLA-DP2 to beryllium oxide (BeO) via oropharyngeal aspiration. As opposed to WT mice, BeO-exposed HLA-DP2 Tg mice developed mononuclear infiltrates in a peribronchovascular distribution that were composed of CD4(+) T cells and included regulatory T (Treg) cells. Beryllium-responsive, HLA-DP2-restricted CD4(+) T cells expressing IFN-γ and IL-2 were present in BeO-exposed HLA-DP2 Tg mice and not in WT mice. Using Be-loaded HLA-DP2-peptide tetramers, we identified Be-specific CD4(+) T cells in the mouse lung that recognize identical ligands as CD4(+) T cells derived from the human lung. Importantly, a subset of HLA-DP2 tetramer-binding CD4(+) T cells expressed forkhead box P3, consistent with the expansion of antigen-specific Treg cells. Depletion of Treg cells in BeO-exposed HLA-DP2 Tg mice exacerbated lung inflammation and enhanced granuloma formation. These findings document, for the first time to our knowledge, the development of a Be-specific adaptive immune response in mice expressing HLA-DP2 and the ability of Treg cells to modulate the beryllium-induced granulomatous immune response.


Subject(s)
Berylliosis/immunology , Disease Models, Animal , Granuloma/immunology , HLA-DP beta-Chains/immunology , Inflammation/immunology , T-Lymphocytes, Regulatory/immunology , Adaptive Immunity/genetics , Adaptive Immunity/immunology , Animals , Berylliosis/genetics , Beryllium/immunology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Enzyme-Linked Immunospot Assay , Flow Cytometry , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Granuloma/genetics , HLA-DP beta-Chains/genetics , Humans , Inflammation/genetics , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-2/immunology , Interleukin-2/metabolism , Lung/immunology , Lung/metabolism , Lung/pathology , Mice , Mice, Transgenic , Spleen/immunology , Spleen/metabolism , Spleen/pathology , T-Lymphocytes, Regulatory/metabolism
7.
Proc Natl Acad Sci U S A ; 110(12): E1122-31, 2013 Mar 19.
Article in English | MEDLINE | ID: mdl-23447566

ABSTRACT

Many vaccines include aluminum salts (alum) as adjuvants despite little knowledge of alum's functions. Host DNA rapidly coats injected alum. Here, we further investigated the mechanism of alum and DNA's adjuvant function. Our data show that DNase coinjection reduces CD4 T-cell priming by i.m. injected antigen + alum. This effect is partially replicated in mice lacking stimulator of IFN genes, a mediator of cellular responses to cytoplasmic DNA. Others have shown that DNase treatment impairs dendritic cell (DC) migration from the peritoneal cavity to the draining lymph node in mice immunized i.p. with alum. However, our data show that DNase does not affect accumulation of, or expression of costimulatory proteins on, antigen-loaded DCs in lymph nodes draining injected muscles, the site by which most human vaccines are administered. DNase does inhibit prolonged T-cell-DC conjugate formation and antigen presentation between antigen-positive DCs and antigen-specific CD4 T cells following i.m. injection. Thus, from the muscle, an immunization site that does not require host DNA to promote migration of inflammatory DCs, alum acts as an adjuvant by introducing host DNA into the cytoplasm of antigen-bearing DCs, where it engages receptors that promote MHC class II presentation and better DC-T-cell interactions.


Subject(s)
Adjuvants, Immunologic/pharmacology , Alum Compounds/pharmacology , Antigen Presentation/drug effects , CD4-Positive T-Lymphocytes/immunology , DNA/immunology , Dendritic Cells/immunology , Histocompatibility Antigens Class II/immunology , Animals , Antigens/immunology , Cell Communication/drug effects , Cell Movement/drug effects , Humans , Mice , Mice, Knockout , Mice, Nude
8.
J Immunol ; 190(3): 913-21, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23275606

ABSTRACT

We re-examined the observation that γδ T cells, when transferred from mice tolerized to an inhaled conventional Ag, suppress the allergic IgE response to this Ag specifically. Using OVA and hen egg lysozyme in crisscross fashion, we confirmed the Ag-specific IgE-regulatory effect of the γδ T cells. Although only Vγ4(+) γδ T cells are regulators, the Ag specificity does not stem from specificity of their γδ TCRs. Instead, the Vγ4(+) γδ T cells failed to respond to either Ag, but rapidly acquired Ag-specific regulatory function in vivo following i.v. injection of non-T cells derived from the spleen of Ag-tolerized mice. This correlated with their in vivo Ag acquisition from i.v. injected Ag-loaded splenic non-T cells, and in vivo transfer of membrane label provided evidence for direct contact between the injected splenic non-T cells and the Vγ4(+) γδ T cells. Together, our data suggest that Ag itself, when acquired by γδ T cells, directs the specificity of their IgE suppression.


Subject(s)
Antigens/immunology , Asthma/immunology , Immunoglobulin E/immunology , Muramidase/immunology , Ovalbumin/immunology , Receptors, Antigen, T-Cell, gamma-delta/analysis , T-Lymphocyte Subsets/immunology , Administration, Inhalation , Adoptive Transfer , Aerosols , Animals , Antigens/administration & dosage , Antigens/toxicity , Asthma/etiology , Cell Separation , Female , Humans , Immune Tolerance , Immunological Synapses , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Muramidase/administration & dosage , Muramidase/toxicity , Ovalbumin/administration & dosage , Ovalbumin/toxicity , Spleen/immunology , T-Cell Antigen Receptor Specificity , T-Lymphocyte Subsets/transplantation
9.
Proc Natl Acad Sci U S A ; 108(19): 7914-9, 2011 May 10.
Article in English | MEDLINE | ID: mdl-21518876

ABSTRACT

Vaccines can greatly reduce the spread of and deaths from many infectious diseases. However, many infections have no successful vaccines. Better understanding of the generation of protective CD8 memory T cells by vaccination is essential for the rational design of new vaccines that aim to prime cellular immune responses. Here we demonstrate that the combination of two adjuvants that are currently licensed for use in humans can be used to prime long-lived memory CD8 T cells that protect mice from viral challenge. The universally used adjuvant, aluminum salts, primed long-lived memory CD8 T cells; however, effective cytotoxic T-cell differentiation occurred only in the presence of an additional adjuvant, monophosphoryl lipid A (MPL). MPL-induced IL-6 was required for cytotoxic differentiation. The IL-6 acted by inducing granzyme B production and reducing expression of inhibitory molecule PD1 on the surface of the primed CD8 T cells. CD8 memory T cells generated by antigen delivered with both aluminum salts and MPL provided significant protection from influenza A challenge. These adjuvants could be used in human vaccines to prime protective memory CD8 T cells.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Alum Compounds/administration & dosage , Lipid A/analogs & derivatives , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Subunit/administration & dosage , Animals , Antigen Presentation , Antigens, Surface/biosynthesis , Apoptosis Regulatory Proteins/biosynthesis , Cattle , Cytokines/biosynthesis , Female , Humans , Immunologic Memory , Influenza A virus/immunology , Interleukin-6/biosynthesis , Interleukin-6/deficiency , Interleukin-6/genetics , Lipid A/administration & dosage , Mice , Mice, Knockout , Mice, Transgenic , Nucleocapsid Proteins , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Ovalbumin/administration & dosage , Ovalbumin/immunology , Programmed Cell Death 1 Receptor , RNA-Binding Proteins/administration & dosage , RNA-Binding Proteins/immunology , Serum Albumin, Bovine/administration & dosage , Serum Albumin, Bovine/immunology , Viral Core Proteins/administration & dosage , Viral Core Proteins/immunology
10.
J Immunol ; 186(5): 2889-96, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21270407

ABSTRACT

CD4 T cell help for B cells is critical for effective Ab responses. Although many of the molecules involved in helper functions of naive CD4 T cells have been characterized, much less is known about the helper capabilities of memory CD4 T cells, an important consideration for the design of vaccines that aim to prime protective memory CD4 T cells. In this study, we demonstrate that memory CD4 T cells enable B cells to expand more rapidly and class switch earlier than do primary responding CD4 T cells. This accelerated response does not require large numbers of memory cells, and similar numbers of primary responding cells provide less effective help than do memory cells. However, only memory CD4 T cells that express the B cell follicle homing molecule, CXCR5, are able to accelerate the response, suggesting that the rapidity of the Ab response depends on the ability of CD4 memory T cells to migrate quickly toward B cells.


Subject(s)
B-Lymphocyte Subsets/immunology , Immunologic Memory , Receptors, CXCR5/biosynthesis , Amino Acid Sequence , Animals , B-Lymphocyte Subsets/microbiology , B-Lymphocyte Subsets/virology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/transplantation , Cell Differentiation/immunology , Cell Movement/immunology , Female , Immunoglobulin G/biosynthesis , Immunoglobulin G/classification , Lymphocyte Activation/immunology , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/pathogenicity , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Receptors, CXCR5/physiology , Receptors, Lymphocyte Homing/administration & dosage , Receptors, Lymphocyte Homing/biosynthesis , Receptors, Lymphocyte Homing/physiology , Resting Phase, Cell Cycle/immunology
11.
Blood ; 116(24): 5191-9, 2010 Dec 09.
Article in English | MEDLINE | ID: mdl-20876456

ABSTRACT

It has been recognized for nearly 80 years that insoluble aluminum salts are good immunologic adjuvants and that they form long-lived nodules in vivo. Nodule formation has long been presumed to be central for adjuvant activity by providing an antigen depot, but the composition and function of these nodules is poorly understood. We show here that aluminum salt nodules formed within hours of injection and contained the clotting protein fibrinogen. Fibrinogen was critical for nodule formation and required processing to insoluble fibrin by thrombin. DNase treatment partially disrupted the nodules, and the nodules contained histone H3 and citrullinated H3, features consistent with extracellular traps. Although neutrophils were not essential for nodule formation, CD11b(+) cells were implicated. Vaccination of fibrinogen-deficient mice resulted in normal CD4 T-cell and antibody responses and enhanced CD8 T-cell responses, indicating that nodules are not required for aluminum's adjuvant effect. Moreover, the ability of aluminum salts to retain antigen in the body, the well-known depot effect, was unaffected by the absence of nodules. We conclude that aluminum adjuvants form fibrin-dependent nodules in vivo, that these nodules have properties of extracellular traps, and the nodules are not required for aluminum salts to act as adjuvants.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Aluminum/administration & dosage , Adjuvants, Immunologic/pharmacology , Aluminum/pharmacology , Animals , Fibrin/metabolism , Fibrinogen/metabolism , Histones/metabolism , Mice , Mice, Knockout , Salts , T-Lymphocytes/immunology , Thrombin/metabolism , Vaccination
12.
J Immunol ; 183(7): 4403-14, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19734227

ABSTRACT

To understand more about how the body recognizes alum we characterized the early innate and adaptive responses in mice injected with the adjuvant. Within hours of exposure, alum induces a type 2 innate response characterized by an influx of eosinophils, monocytes, neutrophils, DCs, NK cells and NKT cells. In addition, at least 13 cytokines and chemokines are produced within 4 h of injection including IL-1beta and IL-5. Optimal production of some of these, including IL-1beta, depends upon both macrophages and mast cells, whereas production of others, such as IL-5, depends on mast cells only, suggesting that both of these cell types can detect alum. Alum induces eosinophil accumulation partly through the production of mast cell derived IL-5 and histamine. Alum greatly enhances priming of endogenous CD4 and CD8 T cells independently of mast cells, macrophages, and of eosinophils. In addition, Ab levels and Th2 bias was similar in the absence of these cells. We found that the inflammation induced by alum was unchanged in caspase-1-deficient mice, which cannot produce IL-1beta. Furthermore, endogenous CD4 and CD8 T cell responses, Ab responses and the Th2 bias were also not impacted by the absence of caspase-1 or NLRP3. These data suggest that activation of the inflammasome and the type 2 innate response orchestrated by macrophages and mast cells in vivo are not required for adjuvant effect of alum on endogenous T and B cell responses.


Subject(s)
Adjuvants, Immunologic/pharmacology , Alum Compounds/pharmacology , Immunity, Cellular/drug effects , Immunity, Innate/drug effects , Inflammation Mediators/pharmacology , Macrophages, Peritoneal/immunology , Mast Cells/immunology , Adjuvants, Immunologic/administration & dosage , Alum Compounds/administration & dosage , Amino Acid Sequence , Animals , Biosensing Techniques , Carrier Proteins/physiology , Caspase 1/physiology , Cell Movement/drug effects , Cell Movement/immunology , Inflammation Mediators/administration & dosage , Inflammation Mediators/classification , Injections, Intraperitoneal , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mast Cells/drug effects , Mast Cells/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , NLR Family, Pyrin Domain-Containing 3 Protein
13.
BMC Biol ; 8: 37, 2010 Apr 12.
Article in English | MEDLINE | ID: mdl-20385031

ABSTRACT

For many diseases vaccines are lacking or only partly effective. Research on protective immunity and adjuvants that generate vigorous immune responses may help generate effective vaccines against such pathogens.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antibody Formation/immunology , Immunity, Cellular/immunology , Signal Transduction/immunology , Vaccination/methods , CD4-Positive T-Lymphocytes/immunology , Humans , Immunologic Memory/immunology
14.
JCI Insight ; 5(9)2020 05 07.
Article in English | MEDLINE | ID: mdl-32255768

ABSTRACT

Chronic beryllium disease (CBD) is a metal hypersensitivity/autoimmune disease in which damage-associated molecular patterns (DAMPs) promote a break in T cell tolerance and expansion of Be2+/self-peptide-reactive CD4+ T cells. In this study, we investigated the mechanism of cell death induced by beryllium particles in alveolar macrophages (AMs) and its impact on DAMP release. We found that phagocytosis of Be led to AM cell death independent of caspase, receptor-interacting protein kinases 1 and 3, or ROS activity. Before cell death, Be-exposed AMs secreted TNF-α that boosted intracellular stores of IL-1α followed by caspase-8-dependent fragmentation of DNA. IL-1α and nucleosomal DNA were subsequently released from AMs upon loss of plasma membrane integrity. In contrast, necrotic AMs released only unfragmented DNA and necroptotic AMs released only IL-1α. In mice exposed to Be, TNF-α promoted release of DAMPs and was required for the mobilization of immunogenic DCs, the expansion of Be-reactive CD4+ T cells, and pulmonary inflammation in a mouse model of CBD. Thus, early autocrine effects of particle-induced TNF-α on AMs led to a break in peripheral tolerance. This potentially novel mechanism may underlie the known relationship between fine particle inhalation, TNF-α, and loss of peripheral tolerance in T cell-mediated autoimmune disease and hypersensitivities.


Subject(s)
Berylliosis/immunology , CD4-Positive T-Lymphocytes , Macrophages, Alveolar , Tumor Necrosis Factor-alpha/physiology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Line , Chronic Disease , Female , Macrophages, Alveolar/immunology , Macrophages, Alveolar/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
15.
Int Immunol ; 20(5): 659-69, 2008 May.
Article in English | MEDLINE | ID: mdl-18343889

ABSTRACT

Alum is used as a vaccine adjuvant and induces T(h)2 responses and T(h)2-driven antibody isotype production against co-injected antigens. Alum also promotes the appearance in the spleen of Gr1+IL-4+ innate cells that, via IL-4 production, induce MHC II-mediated signaling in B cells. To investigate whether these Gr1+ cells accumulate in the spleen in response to other T(h)2-inducing stimuli and to understand some of their functions, the effects of injection of alum and eggs from the helminth, Schistosoma mansoni, were compared. Like alum, schistosome eggs induced the appearance of Gr1+IL-4+ cells in spleen and promoted MHC II-mediated signaling in B cells. Unlike alum, however, schistosome eggs did not promote CD4 T cell responses against co-injected antigens, suggesting that the effects of alum or schistosome eggs on splenic B cells cannot by themselves explain the T cell adjuvant properties of alum. Accordingly, depletion of IL-4 or Gr1+ cells in alum-injected mice had no effect on the ability of alum to improve expansion of primary CD4 T cells. However, Gr1+ cells and IL-4 played some role in the effects of alum, since depletion of either resulted in antibody responses to antigen that included not only the normal T(h)2-driven isotypes, like IgG1, but also a T(h)1-driven isotype, IgG2c. These data suggest that alum affects the immune response in at least two ways: one, independent of Gr1+ cells and IL-4, that promotes CD4 T cell proliferation and another, via Gr1+IL-4+ cells, that participates in the polarization of the response.


Subject(s)
B-Lymphocytes/immunology , Interleukin-4/metabolism , Receptors, Chemokine/metabolism , Th1 Cells/immunology , Th2 Cells/immunology , Adjuvants, Immunologic , Alum Compounds , Animals , Antigens, Helminth/immunology , Antigens, Helminth/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Female , Genes, MHC Class II , Immunity, Innate , Immunoglobulin Isotypes/biosynthesis , Immunoglobulin Isotypes/immunology , Interleukin-4/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Ovalbumin/immunology , Receptors, Chemokine/immunology , Schistosoma mansoni/immunology , Spleen/immunology
16.
JCI Insight ; 52019 05 16.
Article in English | MEDLINE | ID: mdl-31094704

ABSTRACT

Susceptibility to chronic beryllium (Be) disease is linked to HLA-DP molecules possessing a glutamic acid at the 69th position of the ß-chain (ßGlu69), with the most prevalent ßGlu69-containing molecule being HLA-DP2. We have previously shown that HLA-DP2 transgenic (Tg) mice exposed to Be oxide (BeO) develop mononuclear infiltrates in a peribronchovascular distribution and a beryllium-specific, HLA-DP2-restricted CD4+ T cell response. In addition to T cells, B cells constituted a major portion of infiltrated leukocytes in the lung of BeO-exposed HLA-DP2 Tg mice and sequester BeO particles within ectopic lymphoid aggregates and granulomas. B cell depletion was associated with a loss of lymphoid aggregates and granulomas as well as a significant increase in lung injury in BeO-exposed mice. The protective role of B cells was innate in origin, and BeO-induced B cell recruitment to the lung was dependent on MyD88 signaling. Similar to BeO-exposed HLA-DP2 mice, B cells also accumulate in the lungs of CBD subjects, located at the periphery and surrounding the granuloma. Overall, our data suggest a novel modulatory role for B cells in the protection of the lung against sterile particulate exposure, with B cell recruitment to the inflamed lung occurring in an antigen-independent and MyD88-dependent manner.


Subject(s)
B-Lymphocytes/immunology , HLA-DP beta-Chains/metabolism , Lung Injury/immunology , Lung Injury/prevention & control , Adaptive Immunity , Animals , Beryllium , CD4-Positive T-Lymphocytes/immunology , Chemokine CXCL13/metabolism , Chemokines/metabolism , Cytokines/metabolism , Granuloma , Inflammation , Lung/pathology , Lung Injury/pathology , Lymphocytes/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Differentiation Factor 88 , Tertiary Lymphoid Structures/pathology
17.
J Am Heart Assoc ; 8(15): e013111, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31339057

ABSTRACT

Background Inflammation underlies many forms of pulmonary hypertension (PH), including that resulting from Schistosoma infection, a major cause of PH worldwide. Schistosomiasis-associated PH is proximately triggered by embolization of parasite eggs into the lungs, resulting in localized type 2 inflammation. However, the role of CD4+ T cells in this disease is not well defined. Methods and Results We used a mouse model of schistosomiasis-associated PH, induced by intraperitoneal egg sensitization followed by intravenous egg challenge, with outcomes including right ventricle systolic pressure measured by cardiac catheterization, and cell density and phenotype assessed by flow cytometry. We identified that embolization of Schistosoma eggs into lungs of egg-sensitized mice increased the perivascular density of T-helper 2 (Th2) CD4+ T cells by recruitment of cells from the circulation and triggered type 2 inflammation. Parabiosis confirmed that egg embolization is required for localized type 2 immunity. We found Th2 CD4+ T cells were necessary for Schistosoma-induced PH, given that deletion of CD4+ T cells or inhibiting their Th2 function protected against type 2 inflammation and PH following Schistosoma exposure. We also observed that adoptive transfer of Schistosoma-sensitized CD4+ Th2 cells was sufficient to drive type 2 inflammation and PH. Conclusions Th2 CD4+ T cells are a necessary and sufficient component for the type 2 inflammation-induced PH following Schistosoma exposure.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Hypertension, Pulmonary/immunology , Hypertension, Pulmonary/parasitology , Pneumonia/immunology , Pneumonia/parasitology , Schistosomiasis/complications , Schistosomiasis/immunology , Th2 Cells/immunology , Animals , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL
18.
Curr Opin Immunol ; 47: 44-51, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28734174

ABSTRACT

Adjuvants have been deliberately added to vaccines since the 1920's when alum was discovered to boost antibody responses, leading to better protection. The first adjuvants were discovered by accident and were used in the safer but less immunogenic subunit vaccines, supposedly by providing an antigen depot to extend antigen presentation. Since that time, much has been discovered about how these adjuvants impact cells at the tissue site to activate innate immune responses, mobilize dendritic cells and drive adaptive immunity. New approaches to vaccine construction for infectious diseases that have so far not been well addressed by conventional vaccines often attempt to induce antibodies, polyfunctional CD4+ T cells and CD8+ CTLs. The discovery of pattern recognition receptors and ligands that drive desired T cell responses has led to development of novel adjuvant strategies using immunomodulatory agents to direct appropriate immune responses.


Subject(s)
Adjuvants, Immunologic , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Vaccines/immunology , Adaptive Immunity , Alum Compounds/therapeutic use , Animals , Antibody Formation , Antigen Presentation , Humans , Lymphocyte Activation
19.
Curr Opin Immunol ; 42: 25-30, 2016 10.
Article in English | MEDLINE | ID: mdl-27228132

ABSTRACT

Metal-induced hypersensitivity is driven by T cell sensitization to metal ions. Recent advances in our understanding of the complex interactions between innate and adaptive immunity have expanded our knowledge of the pathogenesis of these diseases. Metals activate the innate immune system through direct binding to pathogen recognition receptors, activation of the inflammasome, or the induction of cellular death and release of alarmins. Certain metals can serve as adjuvants, promoting dendritic cell activation and migration as well as antigen presentation to metal-specific T cells. These T cells can recognize metals as haptens or as altered MHC-peptide complexes. The ability of metals to create these neoantigens emphasizes the similarity between metal-induced hypersensitivity and autoimmunity.


Subject(s)
Allergens/immunology , Autoantigens/immunology , CD4-Positive T-Lymphocytes/immunology , Hypersensitivity, Delayed/immunology , Metals/immunology , Adaptive Immunity , Animals , Apoptosis , Autoimmunity , Histocompatibility Antigens Class II/metabolism , Humans , Immunity, Innate , Protein Binding , Receptors, Pattern Recognition/metabolism
20.
Curr Opin Immunol ; 25(6): 775-80, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23978481

ABSTRACT

Chronic beryllium disease (CBD) is a granulomatous lung disorder caused by a hypersensitivity to beryllium and characterized by the accumulation of beryllium-specific CD4(+) T cells in the lung. Genetic susceptibility to beryllium-induced disease is strongly associated with HLA-DP alleles possessing a glutamic acid at the 69th position of the ß-chain (ßGlu69). The structure of HLA-DP2, the most prevalent ßGlu69-containing molecule, revealed a unique solvent-exposed acidic pocket that includes ßGlu69 and represents the putative beryllium-binding site. The delineation of mimotopes and endogenous self-peptides that complete the αßTCR ligand for beryllium-specific CD4(+) T cells suggests a unique role of these peptides in metal ion coordination and the generation of altered self-peptides, blurring the distinction between hypersensitivity and autoimmunity.


Subject(s)
Berylliosis/immunology , CD4-Positive T-Lymphocytes/immunology , Berylliosis/genetics , Beryllium/immunology , Chronic Disease , Genetic Predisposition to Disease , Humans
SELECTION OF CITATIONS
SEARCH DETAIL