Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Mucosal Immunol ; 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39069078

ABSTRACT

Early-life (EL) respiratory infections increase pulmonary disease risk, especially EL-Respiratory Syncytial Virus (EL-RSV) infections linked to asthma. Mechanisms underlying asthma predisposition remain unknown. In this study, we examined the long-term effects on the lung after four weeks post EL-RSV infection. We identified alterations in the lung epithelial cell, with a rise in the percentage of alveolar type 2 epithelial cells (AT2) and a decreased percentage of cells in the AT1 and AT2-AT1 subclusters, as well as upregulation of Bmp2 and Krt8 genes that are associated with AT2-AT1 trans-differentiation, suggesting potential defects in lung repair processes. We identified persistent upregulation of asthma-associated genes, including Il33. EL-RSV-infected mice allergen-challenged exhibited exacerbated allergic response, with significant upregulation of Il33 in the lung and AT2 cells. Similar long-term effects were observed in mice exposed to EL-IL-1ß. Notably, treatment with IL-1ra during acute EL-RSV infection mitigated the long-term alveolar alterations and the allergen-exacerbated response. Finally, epigenetic modifications in the promoter of the Il33 gene were detected in AT2 cells harvested from EL-RSV and EL-IL1ß groups, suggesting that long-term alteration in the epithelium after RSV infection is dependent on the IL-1ß pathway. This study provides insight into the molecular mechanisms of asthma predisposition after RSV infection.

2.
Semin Immunopathol ; 46(3-4): 9, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39066790

ABSTRACT

Allergic diseases affect up to 40% of the global population with a substantial rise in food allergies, in particular, over the past decades. For the majority of individuals with allergy fundamental programming of a pro-allergic immune system largely occurs in early childhood where it is crucially governed by prenatal genetic and environmental factors, including their interactions. These factors include several genetic aberrations, such as filaggrin loss-of-function mutations, early exposure to respiratory syncytial virus, and various chemicals such as plasticizers, as well as the influence of the gut microbiome and numerous lifestyle circumstances. The effects of such a wide range of factors on allergic responses to an array of potential allergens is complex and the severity of these responses in a clinical setting are subsequently not easy to predict at the present time. However, some parameters which condition a pro-allergic immune response, including severe anaphylaxis, are becoming clearer. This review summarises what we currently know, and don't know, about the factors which influence developing pro-allergic immunity particularly during the early-life perinatal period.


Subject(s)
Filaggrin Proteins , Hypersensitivity , Humans , Risk Factors , Hypersensitivity/immunology , Hypersensitivity/etiology , Hypersensitivity/metabolism , Animals , Disease Susceptibility , Pregnancy , Allergens/immunology , Gastrointestinal Microbiome/immunology , Prenatal Exposure Delayed Effects/immunology , Environmental Exposure/adverse effects
3.
J Allergy Clin Immunol ; 152(6): 1607-1618.e1, 2023 12.
Article in English | MEDLINE | ID: mdl-37604310

ABSTRACT

BACKGROUND: IgA is the most abundant immunoglobulin at the mucosal surface and although its role in regulating mucosal immunity is not fully understood, its presence is associated with protection from developing allergic disease. OBJECTIVE: We sought to determine the role of IgA immune complexes for therapeutic application to mucosal allergic responses. METHODS: Trinitrophenol (TNP)-specific IgA immune complexes were applied, using TNP-coupled ovalbumin (OVA), to airway and gut mucosal surfaces in systemically sensitized allergic animals to regulate allergen challenge responses. Animals were assessed for both pathologic and immune-mediated responses in the lung and gut, respectively, using established mouse models. RESULTS: The mucosal application of IgA immune complexes in the lung and gut with TNP-OVA regulated TH2-driven allergic response in the lung and gut, reducing TH2 cytokines and mucus (lung) as well as diarrhea and temperature loss (gut), but increasing IL-10 and the number of regulatory T cells. The IgA-OVA immune complex did not alter peanut-induced anaphylaxis, indicating antigen specificity. Using OVA-specific DO.11-green fluorescent protein IL-4 reporter mouse-derived TH2-skewed cells in a transfer model demonstrated that mucosal IgA immune complex treatment reduced TH2-cell expansion and increased the number of regulatory T cells. To address a potential mechanism of action, TGF-ß and IL-10 were induced in bone marrow-derived dendritic cells when they were exposed to IgA immune complex, suggesting a regulatory phenotype induced in dendritic cells that also led to an altered primary T-cell-mediated response in in vitro OVA-specific assays. CONCLUSIONS: These studies highlight one possible mechanism of how allergen-specific IgA may provide a regulatory signal to reduce the development of allergic responses in the lung and gut.


Subject(s)
Anaphylaxis , Interleukin-10 , Animals , Mice , Interleukin-10/metabolism , Antigen-Antibody Complex/metabolism , Lung , Cytokines/metabolism , Allergens , Anaphylaxis/pathology , Ovalbumin , Th2 Cells , Mice, Inbred BALB C , Disease Models, Animal
4.
Mucosal Immunol ; 16(5): 727-739, 2023 10.
Article in English | MEDLINE | ID: mdl-37557983

ABSTRACT

Eosinophilic esophagitis (EoE) is a T helper (Th)2-mediated inflammatory disorder characterized endoscopically by eosinophilic infiltration leading to fibrosis of the esophagus. Stem cell factor (SCF), a multifunctional cytokine, is upregulated in several allergic diseases, including in patients with EoE. Mast cells and eosinophils express c-kit, the cell surface receptor for SCF, and have been found to play an important role in EoE. Therefore, we investigated whether blocking SCF represents a potential therapeutic approach for EoE. Esophageal inflammation was induced in mice using peanut allergen. In mice with experimental EoE, we found that SCF was upregulated in the esophageal tissue. In EoE mice injected with a polyclonal antibody specific for SCF, we observed a decrease in both mast cells and eosinophils by histological and flow cytometric analysis. Furthermore, Th2 cytokines in the esophagus were decreased in anti-SCF treated mice, as were levels of Th2 cytokines from lung-draining and esophageal lymph nodes. Serum levels of peanut-specific immunoglobulin E were reduced following treatment with anti-SCF. In Kitlf/f-Col1-Cre-ERT mice, which have SCF deleted primarily in myofibroblasts that develop in EoE, we observed similar results as the anti-SCF treated animals for inflammatory cell accumulation, cytokines, and histopathology. These results indicate that therapeutic treatments targeting SCF can reduce allergic inflammation in EoE.


Subject(s)
Eosinophilic Esophagitis , Humans , Mice , Animals , Eosinophilic Esophagitis/drug therapy , Stem Cell Factor/metabolism , Inflammation/metabolism , Disease Models, Animal , Cytokines/metabolism , Eosinophils , Allergens
5.
Front Allergy ; 4: 1105588, 2023.
Article in English | MEDLINE | ID: mdl-37304168

ABSTRACT

In recent years the increased incidence of food allergy in Western culture has been associated with environmental factors and an inappropriate immune phenotype. While the adaptive immune changes in food allergy development and progression have been well-characterized, an increase in innate cell frequency and activation status has also recently received greater attention. Early in prenatal and neonatal development of human immunity there is a reliance on epigenetic and metabolic changes that stem from environmental factors, which are critical in training the immune outcomes. In the present review, we discuss how trained immunity is regulated by epigenetic, microbial and metabolic factors, and how these factors and their impact on innate immunity have been linked to the development of food allergy. We further summarize current efforts to use probiotics as a potential therapeutic approach to reverse the epigenetic and metabolic signatures and prevent the development of severe anaphylactic food allergy, as well as the potential use of trained immunity as a diagnostic and management strategy. Finally, trained immunity is presented as one of the mechanisms of action of allergen-specific immunotherapy to promote tolerogenic responses in allergic individuals.

6.
J Exp Med ; 218(11)2021 11 01.
Article in English | MEDLINE | ID: mdl-34613328

ABSTRACT

Development of the immune system can be influenced by diverse extrinsic and intrinsic factors that influence the risk of disease. Severe early life respiratory syncytial virus (RSV) infection is associated with persistent immune alterations. Previously, our group had shown that adult mice orally supplemented with Lactobacillus johnsonii exhibited decreased airway immunopathology following RSV infection. Here, we demonstrate that offspring of mice supplemented with L. johnsonii exhibit reduced airway mucus and Th2 cell-mediated response to RSV infection. Maternal supplementation resulted in a consistent gut microbiome in mothers and their offspring. Importantly, supplemented maternal plasma and breastmilk, and offspring plasma, exhibited decreased inflammatory metabolites. Cross-fostering studies showed that prenatal Lactobacillus exposure led to decreased Th2 cytokines and lung inflammation following RSV infection, while postnatal Lactobacillus exposure diminished goblet cell hypertrophy and mucus production in the lung in response to airway infection. These studies demonstrate that Lactobacillus modulation of the maternal microbiome and associated metabolic reprogramming enhance airway protection against RSV in neonates.


Subject(s)
Gastrointestinal Microbiome/immunology , Respiratory Syncytial Virus Infections/immunology , Animals , Cytokines/immunology , Disease Models, Animal , Female , Lung/immunology , Lung/virology , Mice , Mice, Inbred BALB C , Pregnancy , Th2 Cells/immunology
7.
Front Immunol ; 12: 604192, 2021.
Article in English | MEDLINE | ID: mdl-33786039

ABSTRACT

Food allergy is a growing public health problem with ~15 million people affected in the United States. In allergic food disease, IgE on mast cells bind to ingested antigens leading to the activation and degranulation of mast cells. Stem cell factor (SCF) is mast cell growth and activation factor that is required for peripheral tissue mast cells. We targeted a specific isoform of SCF, the larger 248 amino acid form, that drives peripheral tissue mast cell differentiation using a specific monoclonal antibody in a model of food allergy. Ovalbumin sensitized and intragastrically challenged mice were monitored for symptoms of anaphylaxis including respiratory distress, diarrhea, and a reduction in body temperature. During the second week of challenges, allergic mice were injected with an antibody to block SCF248 or given IgG control. Mice treated with α-SCF248 had a decreased incidence of diarrhea and no reduction in body temperature suggesting a reduction in anaphylaxis compared to IgG control treated animals. Re-stimulated mesenteric lymph nodes indicated that α-SCF248 treated mice had decreased OVA-specific Th2 cytokine production compared to IgG control treated allergic animals. The reduction of food induced anaphylaxis was accompanied by a significant reduction in gut leak. The mesenteric lymph node cells were analyzed by flow cytometry and showed a decrease in the number of type 2 innate lymphoid cells in mice injected with α-SCF248. Morphometric enumeration of esterase+ mast cells demonstrated a significant reduction throughout the small intestine. Using a more chronic model of persistent food-induced anaphylaxis, short term therapeutic treatment with α-SCF248 during established disease effectively blocked food induced anaphylaxis. Together, these data suggest that therapeutically blocking SCF248 in food allergic animals can reduce the severity of food allergy by reducing mast cell mediated disease activation.


Subject(s)
Anaphylaxis/immunology , Anaphylaxis/prevention & control , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Neutralizing/pharmacology , Food Hypersensitivity/immunology , Stem Cell Factor/antagonists & inhibitors , Allergens/immunology , Anaphylaxis/diagnosis , Anaphylaxis/drug therapy , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/therapeutic use , Biomarkers , Biopsy , Chemokine CCL2/metabolism , Disease Models, Animal , Female , Food Hypersensitivity/diagnosis , Food Hypersensitivity/drug therapy , Immunoglobulin E/immunology , Immunophenotyping , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Mast Cells/drug effects , Mast Cells/immunology , Mast Cells/metabolism , Mice , Th2 Cells/immunology , Th2 Cells/metabolism
8.
J Immunol ; 206(6): 1315-1328, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33514510

ABSTRACT

Our studies have previously shown a role for persistent TSLP production in the lungs of mice after early-life respiratory syncytial virus (RSV) infection that leads to an altered immune phenotype, including accumulation of "inflammatory" dendritic cells (DC). This study investigates the role of TSLP driving systemic trained immunity in DC in early-life RSV-infected mice. Bone marrow-derived DCs (BMDC) from early-life RSV-infected mice at 4 wk postinfection showed enhanced expression of costimulatory molecules and cytokines, including Tslp, that regulate immune cell function. The adoptive transfer of BMDC grown from early-life RSV-infected mice was sufficient to exacerbate allergic disease development. The addition of recombinant TSLP during differentiation of BMDC from naive mice induced a similar altered phenotype as BMDC grown from early-life RSV-infected mice, suggesting a role for TSLP in the phenotypic changes. To assess the role of TSLP in these changes, global transcriptomic characterization of TSLPR-/- BMDC infected with RSV was performed and showed a higher upregulation of type 1 IFN genes and concomitant downregulation of inflammatory genes. Assay for transposase-accessible chromatin using sequencing analysis demonstrated that TSLPR-/- BMDC had a parallel gain in physical chromatin accessibility near type 1 genes and loss in accessibility near genes related to RSV pathology, with IFN regulatory factor 4 (IRF4) and STAT3 predicted as top transcription factors binding within differentially accessible regions in wild-type. Importantly, these studies show that in the absence of TSLP signaling, BMDC are able to mount an appropriate type 1 IFN-associated antiviral response to RSV. In summary, RSV-induced TSLP alters chromatin structure in DC to drive trained innate immunity and activates pathogenic gene programs in mice.


Subject(s)
Chromatin Assembly and Disassembly/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , Age Factors , Animals , Animals, Newborn , Dendritic Cells/metabolism , Disease Models, Animal , Female , Humans , Immunoglobulins/genetics , Immunoglobulins/metabolism , Infant , Interferon Regulatory Factors/metabolism , Interferon Type I/genetics , Male , Mice , Mice, Knockout , Receptors, Cytokine/genetics , Receptors, Cytokine/metabolism , Respiratory Syncytial Virus Infections/genetics , Respiratory Syncytial Virus Infections/virology , STAT3 Transcription Factor/metabolism , Up-Regulation/immunology , Thymic Stromal Lymphopoietin
9.
Mucosal Immunol ; 14(1): 135-143, 2021 01.
Article in English | MEDLINE | ID: mdl-32576925

ABSTRACT

Food-triggered anaphylaxis can encompass a variety of systemic and intestinal symptoms. Murine-based and clinical studies have revealed a role for histamine and H1R and H2R-pathway in the systemic response; however, the molecular processes that regulate the gastrointestinal (GI) response are not as well defined. In the present study, by utilizing an IgE-mast cell (MC)-dependent experimental model of oral antigen-induced anaphylaxis, we define the intestinal epithelial response during a food-induced anaphylactic reaction. We show that oral allergen-challenge stimulates a rapid dysregulation of intestinal epithelial transcellular and paracellular transport that was associated with the development of secretory diarrhea. Allergen-challenge induced (1) a rapid intestinal epithelial Cftr-dependent Cl- secretory response and (2) paracellular macromolecular leak that was associated with modification in epithelial intercellular junction proteins claudin-1, 2, 3 and 5, E-cadherin and desmosomal cadherins. OVA-induced Cftr-dependent Cl- secretion and junctional protein degradation was rapid occurring and was sustained for 72 h following allergen-challenge. Blockade of both the proteolytic activity and Cl- secretory response was required to alleviate intestinal symptoms of food-induced anaphylaxis. Collectively, these data suggest that the GI symptom of food-induced anaphylactic reaction, secretory diarrhea, is a consequence of CFTR-dependent Cl- secretion and proteolytic activity.


Subject(s)
Anaphylaxis/etiology , Anaphylaxis/metabolism , Chlorides/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Food Hypersensitivity/etiology , Food Hypersensitivity/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Allergens/immunology , Anaphylaxis/pathology , Animals , Biomarkers , Disease Models, Animal , Disease Susceptibility , Food Hypersensitivity/pathology , Immunoglobulin E/immunology , Ion Transport , Mast Cells/immunology , Mast Cells/metabolism , Mice
10.
Allergy ; 75(9): 2279-2293, 2020 09.
Article in English | MEDLINE | ID: mdl-32277487

ABSTRACT

BACKGROUND: Respiratory syncytial virus (RSV) affects most infants early in life and is associated with increased asthma risk. The specific mechanism remains unknown. OBJECTIVE: To investigate the role of uric acid (UA) and IL-1ß in RSV immunopathology and asthma predisposition. METHODS: Tracheal aspirates from human infants with and without RSV were collected and analyzed for pro-IL-1ß mRNA and protein to establish a correlation in human disease. Neonatal mouse models of RSV were employed, wherein mice infected at 6-7 days of life were analyzed at 8 days postinfection, 5 weeks postinfection, or after a chronic cockroach allergen asthma model. A xanthine oxidase inhibitor or IL-1 receptor antagonist was administered during RSV infection. RESULTS: Human tracheal aspirates from RSV-infected infants showed elevated pro-IL-1ß mRNA and protein. Inhibition of UA or IL-1ß during neonatal murine RSV infection decreased mucus production, reduced cellular infiltrates to the lung (especially ILC2s), and decreased type 2 immune responses. Inhibition of either UA or IL-1ß during RSV infection led to chronic reductions in pulmonary immune cell composition and reduced type 2 immune responses and reduced similar responses after challenge with cockroach antigen. CONCLUSIONS: Inhibiting UA and IL-1ß during RSV infection ameliorates RSV immunopathology, reduces the consequences of allergen-induced asthma, and presents new therapeutic targets to reduce early-life viral-induced asthma development.


Subject(s)
Asthma , Respiratory Syncytial Virus Infections , Animals , Immunity, Innate , Lung , Lymphocytes , Mice , Mice, Inbred BALB C , Respiratory Syncytial Viruses , Uric Acid
11.
J Immunol ; 204(1): 159-168, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31748348

ABSTRACT

Severe disease following respiratory syncytial virus (RSV) infection has been linked to enhanced proinflammatory cytokine production that promotes a Th2-type immune environment. Epigenetic regulation in immune cells following viral infection plays a role in the inflammatory response and may result from upregulation of key epigenetic modifiers. In this study, we show that RSV-infected bone marrow-derived dendritic cells (BMDC) as well as pulmonary dendritic cells (DC) from RSV-infected mice upregulated the expression of Kdm6b/Jmjd3 and Kdm6a/Utx, H3K27 demethylases. KDM6-specific chemical inhibition (GSK J4) in BMDC led to decreased production of chemokines and cytokines associated with the inflammatory response during RSV infection (i.e., CCL-2, CCL-3, CCL-5, IL-6) as well as decreased MHC class II and costimulatory marker (CD80/86) expression. RSV-infected BMDC treated with GSK J4 altered coactivation of T cell cytokine production to RSV as well as a primary OVA response. Airway sensitization of naive mice with RSV-infected BMDCs exacerbate a live challenge with RSV infection but was inhibited when BMDCs were treated with GSK J4 prior to sensitization. Finally, in vivo treatment with the KDM6 inhibitor, GSK J4, during RSV infection reduced inflammatory DC in the lungs along with IL-13 levels and overall inflammation. These results suggest that KDM6 expression in DC enhances proinflammatory innate cytokine production to promote an altered Th2 immune response following RSV infection that leads to more severe immunopathology.


Subject(s)
Histone Demethylases/immunology , Inflammation/immunology , Respiratory Syncytial Virus Infections/immunology , Up-Regulation , Animals , Cell Line, Tumor , Dendritic Cells/immunology , Dendritic Cells/pathology , Female , Humans , Inflammation/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Respiratory Syncytial Virus Infections/pathology
12.
J Allergy Clin Immunol ; 145(1): 239-254.e2, 2020 01.
Article in English | MEDLINE | ID: mdl-31647967

ABSTRACT

BACKGROUND: The pathology of eosinophilic esophagitis (EoE) is characterized by eosinophil-rich inflammation, basal zone hyperplasia (BZH), and dilated intercellular spaces, and the underlying processes that drive the pathologic manifestations of the disease remain largely unexplored. OBJECTIVE: We sought to investigate the involvement of the calcium-activated chloride channel anoctamin 1 (ANO1) in esophageal proliferation and the histopathologic features of EoE. METHODS: We examined mRNA and protein expression of ANO1 in esophageal biopsy samples from patients with EoE and in mice with EoE. We performed molecular and cellular analyses and ion transport assays on an in vitro esophageal epithelial 3-dimensional model system (EPC2-ALI) and murine models of EoE to define the relationship between expression and function of ANO1 and esophageal epithelial proliferation in patients with EoE. RESULTS: We observed increased ANO1 expression in esophageal biopsy samples from patients with EoE and in mice with EoE. ANO1 was expressed within the esophageal basal zone, and expression correlated positively with disease severity (eosinophils/high-power field) and BZH. Using an in vitro esophageal epithelial 3-dimensional model system revealed that ANO1 undergoes chromatin modification and rapid upregulation of expression after IL-13 stimulation, that ANO1 is the primary apical IL-13-induced Cl- transport mechanism within the esophageal epithelium, and that loss of ANO1-dependent Cl- transport abrogated esophageal epithelial proliferation. Mechanistically, ANO1-dependent regulation of basal cell proliferation was associated with modulation of TP63 expression and phosphorylated cyclin-dependent kinase 2 levels. CONCLUSIONS: These data identify a functional role for ANO1 in esophageal cell proliferation and BZH in patients with EoE and provide a rationale for pharmacologic intervention of ANO1 function in patients with EoE.


Subject(s)
Anoctamin-1/immunology , Eosinophilic Esophagitis/immunology , Epithelial Cells/immunology , Esophagus/immunology , Gene Expression Regulation , Neoplasm Proteins/immunology , Animals , Cell Line , Disease Models, Animal , Eosinophilic Esophagitis/pathology , Epithelial Cells/pathology , Esophagus/pathology , Female , Humans , Male , Mice , Mice, Inbred BALB C
13.
Immunol Allergy Clin North Am ; 39(3): 309-319, 2019 08.
Article in English | MEDLINE | ID: mdl-31284922

ABSTRACT

The infant's developing immune response is central to establishing a balanced system that reacts appropriately to infectious stimuli, but does not induce altered disease states with potential long-term sequelae. Respiratory syncytial virus may alter the immune system, affecting future responses. Early infection may have direct effects on the lung itself. Other early life processes contribute to the development of immune responses including assembly of the microbiome, which seems to have a particularly important role for establishing the immune environment. This review covers studies that have set up important paradigms and discusses recent data that direct research toward informative hypotheses.


Subject(s)
Asthma/etiology , Respiratory Syncytial Virus Infections/complications , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses , Adaptive Immunity/genetics , Age Factors , Animals , Asthma/epidemiology , Disease Susceptibility , Energy Metabolism , Epigenesis, Genetic , Gastrointestinal Microbiome/immunology , Humans , Immunity, Innate/genetics , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/virology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology
14.
Mucosal Immunol ; 12(4): 969-979, 2019 07.
Article in English | MEDLINE | ID: mdl-31076663

ABSTRACT

Many studies have linked severe RSV infection during early-life with an enhanced likelihood of developing childhood asthma, showing a greater susceptibility in boys. Our studies show that early-life RSV infection leads to differential long-term effects based upon the sex of the neonate; leaving male mice prone to exacerbation upon secondary allergen exposure while overall protecting female mice. During initial viral infection, we observed better viral control in the female mice with correlative expression of interferon-ß that was not observed in male mice. Additionally, we observed persistent immune alterations in male mice at 4 weeks post infection. These alterations include Th2 and Th17-skewing, innate cytokine expression (Tslp and Il33), and infiltration of innate immune cells (DC and ILC2). Upon exposure to allergen, beginning at 4 weeks following early-life RSV-infection, male mice show severe allergic exacerbation while female mice appear to be protected. Due to persistent expression of TSLP following early-life RSV infection in male mice, genetically modified TSLPR-/- mice were evaluated and demonstrated an abrogation of allergen exacerbation in male mice. These data indicate that TSLP is involved in the altered immune environment following neonatal RSV-infection that leads to more severe responses in males during allergy exposure, later in life. Thus, TSLP may be a clinically relevant therapeutic target early in life.


Subject(s)
Cytokines/metabolism , Hypersensitivity/immunology , Hypersensitivity/metabolism , Immunomodulation , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Viruses/immunology , Animals , Disease Susceptibility , Female , Hypersensitivity/pathology , Immunoglobulins/metabolism , Immunohistochemistry , Male , Mice , Receptors, Cytokine/metabolism , Respiratory Syncytial Virus Infections/pathology , Sex Factors , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Thymic Stromal Lymphopoietin
15.
Mucosal Immunol ; 12(2): 445-456, 2019 03.
Article in English | MEDLINE | ID: mdl-30617299

ABSTRACT

Stem cell factor (SCF) binds to the receptor c-Kit that is expressed on a number of myeloid and lymphoid cell populations, including Type 2 innate lymphoid cells (ILC2). However the importance of the SCF/c-Kit interaction in ILC2 has not been studied. Here we investigate the role of a specific SCF isoform, SCF248, in the allergic asthmatic response and SCF/c-Kit in ILC2 activation during chronic allergy. We observed that mice treated with a monoclonal antibody specific for SCF248 attenuated the development of chronic asthmatic disease by decreasing the number of mast cells, ILC2 and eosinophils, as well as reducing the accompanying pathogenic cytokine responses. These data were supported using SCFfl/fl-Col1-Cre-ERT mice and W/Wv mice that demonstrated the importance of the stem cell factor/c-Kit activation during chronic allergy and the accumulation of c-kit+ cells. Finally, these data demonstrate for the first time that SCF could activate ILC2 cells in vitro for the production of key allergic cytokines. Together these findings indicate that SCF is a critical cytokine involved in the activation of ILC2 that lead to more severe outcomes during chronic allergy and that the SCF248 isoform could be an important therapeutic target to control the disease progression.


Subject(s)
Asthma/immunology , Lung/pathology , Lymphocytes/immunology , Stem Cell Factor/metabolism , Allergens/immunology , Animals , Cells, Cultured , Chronic Disease , Collagen Type I/genetics , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , Protein Isoforms/genetics , Proto-Oncogene Proteins c-kit/metabolism , Stem Cell Factor/genetics , Stem Cell Factor/immunology , Th2 Cells/immunology
16.
Proc Natl Acad Sci U S A ; 115(45): E10605-E10614, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30348760

ABSTRACT

Hox5 genes (Hoxa5, Hoxb5, Hoxc5) are exclusively expressed in the lung mesenchyme during embryogenesis, and the most severe phenotypes result from constitutive loss of function of all three genes. Because Hox5 triple null mutants exhibit perinatal lethality, the contribution of this paralogous group to postembryonic lung development is unknown. Intriguingly, expression of all three Hox5 genes peaks during the first 2 weeks after birth, reaching levels far exceeding those measured at embryonic stages, and surviving Hoxa5 single and Hox5 AabbCc compound mutants exhibit defects in the localization of alveolar myofibroblasts. To define the contribution of the entire Hox5 paralogous group to this process, we generated an Hoxa5 conditional allele to use with our existing null alleles for Hoxb5 and Hoxc5 Postnatally, mesenchymal deletion of Hoxa5 in an Hoxb5/Hoxc5 double-mutant background results in severe alveolar simplification. The elastin network required for alveolar formation is dramatically disrupted in Hox5 triple mutants, while the basal lamina, interstitial matrix, and fibronectin are normal. Alveolar myofibroblasts remain Pdgfrα+/SMA+ double positive and present in normal numbers, indicating that the irregular elastin network is not due to fibroblast differentiation defects. Rather, we observe that SMA+ myofibroblasts of Hox5 triple mutants are morphologically abnormal both in vivo and in vitro with highly reduced adherence to fibronectin. This loss of adhesion is a result of loss of the integrin heterodimer Itga5b1 in mutant fibroblasts. Collectively, these data show an important role for Hox5 genes in lung fibroblast adhesion necessary for proper elastin network formation during alveologenesis.


Subject(s)
Cell Adhesion , Elastin/metabolism , Genes, Homeobox , Myofibroblasts/cytology , Pulmonary Alveoli/cytology , Alleles , Animals , Dimerization , Gene Expression Regulation, Developmental , Integrin alpha5/metabolism , Integrin beta1/metabolism , Mice , Mutation , Myofibroblasts/metabolism , Pulmonary Alveoli/metabolism
17.
Front Immunol ; 9: 226, 2018.
Article in English | MEDLINE | ID: mdl-29515570

ABSTRACT

Respiratory syncytial virus (RSV) is a common pathogen that infects virtually all children by 2 years of age and is the leading cause of hospitalization of infants worldwide. While most children experience mild symptoms, some children progress to severe lower respiratory tract infection. Those children with severe disease have a much higher risk of developing childhood wheezing later in life. Many risk factors are known to result in exacerbated disease, including premature birth and early age of RSV infection, when the immune system is relatively immature. The development of the immune system before and after birth may be altered by several extrinsic and intrinsic factors that could lead to severe disease predisposition in children who do not exhibit any currently known risk factors. Recently, the role of the microbiome and the resulting metabolite profile has been an area of intense study in the development of lung disease, including viral infection and asthma. This review explores both known risk factors that can lead to severe RSV-induced disease as well as emerging topics in the development of immunity to RSV and the long-term consequences of severe infection.


Subject(s)
Epigenesis, Genetic/immunology , Microbiota/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus, Human/immunology , Child , Genetic Predisposition to Disease , Genotype , Humans , Immunity, Innate , Incidence , Respiratory Syncytial Virus Infections/diagnosis , Respiratory Syncytial Virus Infections/epidemiology , Respiratory Syncytial Virus Infections/virology , Risk Factors , Severity of Illness Index
18.
J Immunol ; 199(2): 501-509, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28576978

ABSTRACT

Allergic asthma is a significant health burden in western countries, and continues to increase in prevalence. Th2 cells contribute to the development of disease through release of the cytokines IL-4, IL-5, and IL-13, resulting in increased airway eosinophils and mucus hypersecretion. The molecular mechanisms behind the disease pathology remain largely unknown. In this study we investigated a potential regulatory role for the Hox5 gene family, Hoxa5, Hoxb5, and Hoxc5, genes known to be important in lung development within mesenchymal cell populations. We found that Hox5-mutant mice show exacerbated pathology compared with wild-type controls in a chronic allergen model, with an increased Th2 response and exacerbated lung tissue pathology. Bone marrow chimera experiments indicated that the observed enhanced pathology was mediated by immune cell function independent of mesenchymal cell Hox5 family function. Examination of T cells grown in Th2 polarizing conditions showed increased proliferation, enhanced Gata3 expression, and elevated production of IL-4, IL-5, and IL-13 in Hox5-deficient T cells compared with wild-type controls. Overexpression of FLAG-tagged HOX5 proteins in Jurkat cells demonstrated HOX5 binding to the Gata3 locus and decreased Gata3 and IL-4 expression, supporting a role for HOX5 proteins in direct transcriptional control of Th2 development. These results reveal a novel role for Hox5 genes as developmental regulators of Th2 immune cell function that demonstrates a redeployment of mesenchyme-associated developmental genes.


Subject(s)
Allergens/immunology , GATA3 Transcription Factor/genetics , Gene Expression Regulation , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Inflammation/immunology , Th2 Cells/immunology , Animals , Cell Proliferation , GATA3 Transcription Factor/metabolism , Hedgehog Proteins/deficiency , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Interleukin-13/biosynthesis , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-4/biosynthesis , Interleukin-4/genetics , Interleukin-4/immunology , Interleukin-5/biosynthesis , Interleukin-5/genetics , Interleukin-5/immunology , Jurkat Cells , Lung/immunology , Lung/pathology , Lung/physiology , Mesoderm/cytology , Mice , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Binding , T-Lymphocytes/immunology , T-Lymphocytes/physiology , Th2 Cells/metabolism , Transcription Factors
19.
J Leukoc Biol ; 98(4): 601-13, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26059830

ABSTRACT

It is well established that the cytokine IL-12 and the transcription factor STAT4, an essential part of the IL-12 signaling pathway, are critical components of the Th1 differentiation process in T cells. In response to pathogenic stimuli, this process causes T cells to proliferate rapidly and secrete high amounts of the cytokine IFN-γ, leading to the Th1 proinflammatory phenotype. However, there are still unknown components of this differentiation pathway. We here demonstrated that the expression of the histone methyltransferase Mll1 is driven by IL-12 signaling through STAT4 in humans and mice and is critical for the proper differentiation of a naïve T cell to a Th1 cell. Once MLL1 is up-regulated by IL-12, it regulates the proliferation of Th1 cells. As evidence of this, we show that Th1 cells from Mll1(+/-) mice are unable to proliferate rapidly in a Th1 environment in vitro and in vivo. Additionally, upon restimulation with cognate antigen Mll1(+/-), T cells do not convert to a Th1 phenotype, as characterized by IFN-γ output. Furthermore, we observed a reduction in IFN-γ production and proliferation in human peripheral blood stimulated with tetanus toxoid by use of a specific inhibitor of the MLL1/menin complex. Together, our results demonstrate that the MLL1 gene plays a previously unrecognized but essential role in Th1 cell biology and furthermore, describes a novel pathway through which Mll1 expression is regulated.


Subject(s)
Cell Proliferation , Epigenesis, Genetic , Histone-Lysine N-Methyltransferase/immunology , Interleukin-12/immunology , Lymphocyte Activation/immunology , Myeloid-Lymphoid Leukemia Protein/immunology , Th1 Cells/immunology , Adolescent , Adult , Aged , Animals , Cell Differentiation/immunology , Cell Proliferation/genetics , Cells, Cultured , Chromatin Immunoprecipitation , DNA Methylation/genetics , DNA Methylation/immunology , Epigenesis, Genetic/genetics , Epigenesis, Genetic/immunology , Female , Flow Cytometry , Histone-Lysine N-Methyltransferase/biosynthesis , Histone-Lysine N-Methyltransferase/genetics , Humans , Lymphocyte Activation/genetics , Male , Mice , Mice, Knockout , Middle Aged , Myeloid-Lymphoid Leukemia Protein/biosynthesis , Myeloid-Lymphoid Leukemia Protein/genetics , STAT4 Transcription Factor , Th1 Cells/cytology , Young Adult
20.
PLoS Pathog ; 11(6): e1004978, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26083387

ABSTRACT

Respiratory syncytial virus (RSV) infection can result in severe disease partially due to its ability to interfere with the initiation of Th1 responses targeting the production of type I interferons (IFN) and promoting a Th2 immune environment. Epigenetic modulation of gene transcription has been shown to be important in regulating inflammatory pathways. RSV-infected bone marrow-derived DCs (BMDCs) upregulated expression of Kdm5b/Jarid1b H3K4 demethylase. Kdm5b-specific siRNA inhibition in BMDC led to a 10-fold increase in IFN-ß as well as increases in IL-6 and TNF-α compared to control-transfected cells. The generation of Kdm5bfl/fl-CD11c-Cre+ mice recapitulated the latter results during in vitro DC activation showing innate cytokine modulation. In vivo, infection of Kdm5bfl/fl-CD11c-Cre+ mice with RSV resulted in higher production of IFN-γ and reduced IL-4 and IL-5 compared to littermate controls, with significantly decreased inflammation, IL-13, and mucus production in the lungs. Sensitization with RSV-infected DCs into the airways of naïve mice led to an exacerbated response when mice were challenged with live RSV infection. When Kdm5b was blocked in DCs with siRNA or DCs from Kdm5bfl/fl-CD11c-CRE mice were used, the exacerbated response was abrogated. Importantly, human monocyte-derived DCs treated with a chemical inhibitor for KDM5B resulted in increased innate cytokine levels as well as elicited decreased Th2 cytokines when co-cultured with RSV reactivated CD4+ T cells. These results suggest that KDM5B acts to repress type I IFN and other innate cytokines to promote an altered immune response following RSV infection that contributes to development of chronic disease.


Subject(s)
Cytokines/biosynthesis , DNA-Binding Proteins/immunology , Dendritic Cells/immunology , Gene Expression Regulation/immunology , Jumonji Domain-Containing Histone Demethylases/immunology , Respiratory Syncytial Virus Infections/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Chromatin Immunoprecipitation , Coculture Techniques , Cytokines/immunology , Disease Models, Animal , Immunity, Innate/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Respiratory Syncytial Viruses/immunology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL