Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
1.
J Allergy Clin Immunol ; 151(4): 1137-1142.e4, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36403663

RESUMO

BACKGROUND: Deleterious variation in the epidermal differentiation complex (EDC) on chromosome 1 is a well-known genetic determinant of atopic dermatitis (AD) and has been associated with risk of peanut allergy (PA) in population-based studies. OBJECTIVE: Our aim was to determine the effect of genetic variation in the EDC on AD trajectory and risk of PA in early life. METHODS: Genome sequencing was used to measure genetic variation in the EDC in the Learning Early about Peanut Allergy (LEAP) study participants. Association tests were done to identify gene- and variant-level predicted deleterious variation associated with AD severity by using the Scoring Atopic Dermatitis (SCORAD) tool (n = 559) at baseline and each follow-up visit, as well as PA and food allergy in peanut avoiders (n = 275). Predicted deleterious variants included missense variants that were frameshift insertions, frameshift deletions, stop-gain mutations, or stop-loss mutations. Associations between variant load, SCORAD score, and PA were tested by using linear and generalized linear regression models. RESULTS: The genes FLG, FLG2, HRNR, and TCHH1 harbored the most predicted deleterious variation (30, 6, 3, and 1 variant, respectively). FLG variants were associated with SCORAD score at all time points; 4 variants (R1798X, R501X, S126X, and S761fs) drove the association with SCORAD score at each time point, and higher variant load was associated with greater AD severity over time. There was an association between these variants and PA, which remained significant independent of baseline AD severity (odds ratio = 2.63 [95% CI = 1.11-6.01] [P = .02]). CONCLUSIONS: Variation in FLG predicted to be deleterious is associated with AD severity at baseline and longitudinally and has an association with PA independent of baseline severity.


Assuntos
Dermatite Atópica , Hipersensibilidade a Amendoim , Humanos , Hipersensibilidade a Amendoim/genética , Dermatite Atópica/genética , Mutação da Fase de Leitura , Mutação , Arachis/genética
2.
J Allergy Clin Immunol ; 151(4): 841-847, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36732171

RESUMO

Examining the genetics of peanut allergy (PA) in the context of clinical trial interventions and outcomes provides an opportunity to not only understand gene-environment interactions for PA risk but to also understand the benefit of allergen immunotherapy. A consistent theme in the genetics of food allergy is that in keeping with the dual allergen exposure hypothesis, barrier- and immune-related genes are most commonly implicated in food allergy and tolerance. With a focus on PA, we review how genetic risk factors across 3 genes (FLG, MALT1, and HLA-DQA1) have helped delineate distinct allergic characteristics and outcomes in the context of environmental interventions in the Learning Early about Peanut Allergy (LEAP) study and other clinical trials. We specifically consider and present a framework for genetic risk prediction for the development of PA and discuss how genetics, age, and oral consumption intertwine to predict PA outcome. Although there is some promise in this proposed framework, a better understanding of the mechanistic pathways by which PA develops and persists is needed to develop targeted therapeutics for established disease. Only by understanding the mechanisms by which PA develops, persists, and resolves can we identify adjuvants to oral immunotherapy to make older children and adults immunologically similar to their younger, more malleable counterparts and thus more likely to achieve long-term tolerance.


Assuntos
Hipersensibilidade Alimentar , Hipersensibilidade a Amendoim , Criança , Adulto , Humanos , Adolescente , Hipersensibilidade a Amendoim/genética , Hipersensibilidade a Amendoim/terapia , Alérgenos , Fatores de Risco , Hipersensibilidade Alimentar/etiologia , Dessensibilização Imunológica/efeitos adversos , Arachis/genética
3.
Immunity ; 41(1): 141-51, 2014 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-25017467

RESUMO

Immunoglobulin E (IgE) antibodies are known for triggering immediate hypersensitivity reactions such as food anaphylaxis. In this study, we tested whether they might additionally function to amplify nascent antibody and T helper 2 (Th2) cell-mediated responses to ingested proteins and whether blocking IgE would modify sensitization. By using mice harboring a disinhibited form of the IL-4 receptor, we developed an adjuvant-free model of peanut allergy. Mast cells and IgE were required for induction of antibody and Th2-cell-mediated responses to peanut ingestion and they impaired regulatory T (Treg) cell induction. Mast-cell-targeted genetic deletion of the FcεRI signaling kinase Syk or Syk blockade also prevented peanut sensitization. In mice with established allergy, Syk blockade facilitated desensitization and induction of Treg cells, which suppressed allergy when transferred to naive recipients. Our study suggests a key role for IgE in driving Th2 cell and IgE responses while suppressing Treg cells in food allergy.


Assuntos
Imunoglobulina E/imunologia , Hipersensibilidade a Amendoim/imunologia , Receptores de IgE/imunologia , Linfócitos T Reguladores/imunologia , Células Th2/imunologia , Alérgenos/imunologia , Animais , Dessensibilização Imunológica , Modelos Animais de Doenças , Imunoglobulina E/genética , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/genética , Ativação Linfocitária/imunologia , Mastócitos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hipersensibilidade a Amendoim/genética , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/genética , Receptores de IgE/antagonistas & inibidores , Receptores de IgE/genética , Receptores de Interleucina-4/genética , Receptores de Interleucina-4/imunologia , Transdução de Sinais/imunologia , Quinase Syk
4.
J Allergy Clin Immunol ; 149(1): 262-274, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34051223

RESUMO

BACKGROUND: The etiology of food allergy is poorly understood; mouse models are powerful systems to discover immunologic pathways driving allergic disease. C3H/HeJ mice are a widely used model for the study of peanut allergy because, unlike C57BL/6 or BALB/c mice, they are highly susceptible to oral anaphylaxis. However, the immunologic mechanism of this strain's susceptibility is not known. OBJECTIVE: We aimed to determine the mechanism underlying the unique susceptibility to anaphylaxis in C3H/HeJ mice. We tested the role of deleterious Toll-like receptor 4 (Tlr4) or dedicator of cytokinesis 8 (Dock8) mutations in this strain because both genes have been associated with food allergy. METHODS: We generated C3H/HeJ mice with corrected Dock8 or Tlr4 alleles and sensitized and challenged them with peanut. We then characterized the antibody response to sensitization, anaphylaxis response to both oral and systemic peanut challenge, gut microbiome, and biomarkers of gut permeability. RESULTS: In contrast to C3H/HeJ mice, C57BL/6 mice were resistant to anaphylaxis after oral peanut challenge; however, both strains undergo anaphylaxis with intraperitoneal challenge. Restoring Tlr4 or Dock8 function in C3H/HeJ mice did not protect from anaphylaxis. Instead, we discovered enhanced gut permeability resulting in ingested allergens in the bloodstream in C3H/HeJ mice compared to C57BL/6 mice, which correlated with an increased number of goblet cells in the small intestine. CONCLUSIONS: Our work highlights the potential importance of gut permeability in driving anaphylaxis to ingested food allergens; it also indicates that genetic loci outside of Tlr4 and Dock8 are responsible for the oral anaphylactic susceptibility of C3H/HeJ mice.


Assuntos
Mucosa Intestinal/metabolismo , Anafilaxia Cutânea Passiva , Hipersensibilidade a Amendoim/metabolismo , Administração Oral , Animais , Arachis/imunologia , Modelos Animais de Doenças , Feminino , Microbioma Gastrointestinal , Predisposição Genética para Doença , Fatores de Troca do Nucleotídeo Guanina/genética , Masculino , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Mutação , Anafilaxia Cutânea Passiva/genética , Hipersensibilidade a Amendoim/genética , Hipersensibilidade a Amendoim/microbiologia , Permeabilidade , Especificidade da Espécie , Receptor 4 Toll-Like/genética
5.
Clin Exp Allergy ; 52(5): 646-657, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35108754

RESUMO

BACKGROUND: Understanding risk factors for peanut allergy (PA) is essential to develop effective preventive measures. OBJECTIVE: The objective was to ascertain associates and predictors of PA, and the relationship between PA and asthma severity. METHODS: In a population-based birth cohort, we investigated the association between objectively confirmed PA with early-life environmental exposures, filaggrin (FLG)-loss-of-function mutations and other atopic disease. We then examined the association of PA with longitudinal trajectories of sensitization, wheeze and allergic comorbidities, which were previously derived using machine learning. Finally, we ascertained the relationship between PA and asthma severity. RESULTS: PA was confirmed in 30/959 participants with evaluable data. In the multivariate analysis, eczema in infancy (OR = 4.4, 95% CI 1.5-13.2, p = 0.007), egg sensitization at age 3 years (OR = 9.7, 95% CI 3.3-29.9, p < 0.001) and early-life cat ownership (OR = 3.0, 95% CI 1.1-8.4, p = 0.04) were independent associates of PA. In the stratified analysis among 700 participants with genetic information, in children with early-life eczema there was no difference in FLG mutations between children with and without PA (3/18 [16.7%] vs. 42/220 [19.1%], p = 1.00). In contrast, among children without eczema, those with PA were almost eight times more likely to have FLG mutations (2/6 [33.3%] vs. 27/456 [5.9%], p = 0.049). We observed associations between PA and multiple allergic sensitization profiles derived using machine learning, with ~60-fold increase in risk among individuals assigned to multiple early sensitization. PA was significantly associated with persistent wheeze (but not other wheeze phenotypes), and with trajectories of atopic disease characterized by co-morbid persistent eczema and wheeze (but not with transient phenotypes). Children with PA were more likely to have asthma, but among asthmatics we found no evidence of an association between PA and asthma severity. CONCLUSIONS: Peanut allergy is associated with multiple IgE sensitization and early-onset persistent eczema and wheeze. FLG loss-of-function mutations were associated with peanut allergy in children without eczema.


Assuntos
Asma , Eczema , Hipersensibilidade a Amendoim , Asma/etiologia , Asma/genética , Coorte de Nascimento , Estudos de Coortes , Eczema/complicações , Eczema/epidemiologia , Eczema/genética , Humanos , Lactente , Proteínas de Filamentos Intermediários/genética , Hipersensibilidade a Amendoim/complicações , Hipersensibilidade a Amendoim/epidemiologia , Hipersensibilidade a Amendoim/genética , Sons Respiratórios/genética , Fatores de Risco
6.
Clin Immunol ; 230: 108820, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34365017

RESUMO

BACKGROUND: Peanut oral immunotherapy (POIT) has provided desensitization to peanut allergic individuals. Limited immunological evaluation exists during the first 24-weeks of POIT. OBJECTIVE: Regulatory T-cells (Tregs) are antigen induced immunosuppressive T-cells important in establishing tolerance. Delineation of early immunologic changes contributing to the development of peanut desensitization would help clarify the mechanism of action in POIT. We performed single-cell RNA sequencing (scRNAseq) on Tregs in pediatric subjects undergoing POIT during the first 24-weeks of therapy to evaluate early immunological changes induced by POIT. METHODS: PBMC samples from peanut allergic subjects between 5 and 12 years of age enrolled in a Phase 1/2a POIT study were collected and analyzed at 0, 6, and 24-weeks after POIT initiation and samples were compared to healthy non-peanut allergic controls. Tregs were enriched from PBMCs and scRNAseq analysis performed. Cell Ranger 3.1.0 (10× Genomics) was utilized to identify cell clusters and differentially expressed genes, and results were analyzed with Seurat suite version 3.0.0. RESULTS: Gene analysis revealed 10 major clusters corresponding to different cell types observed to change during POIT when compared to the healthy, non-peanut-allergic state. scRNAseq analysis of Tregs revealed strong CD3G expression correlating with gdTregs. scRNAseq analysis of gdTregs revealed dynamic changes occurring within the first 6-weeks of treatment and cell frequencies of naïve and memory gdTregs at 24-weeks of treatment reducing to levels similar to healthy controls. Analysis of transcriptomic cell identity analysis using SingleR showed gene expression in gdTregs similar to healthy control after 24-weeks of POIT treatment. scRNAseq analysis revealed alterations in gene expression for memory and naïve gdTregs during this timeframe. Specifically, expression of OX40R (TNFRSF4), GITR (TNFRSF18), TGFB1, CTLA4, ISG20, CD69 were upregulated in memory gdTregs compared to naive gdTregs by 24-weeks of POIT, while IL7R and SELL were downregulated in memory gdTregs compared to naïve gdTregs. CONCLUSIONS: There are specific expression profiles of peripheral naïve and mature gdTreg cells in peanut allergic patients undergoing POIT in the first 24-weeks of treatment implicating pathways involved in maintenance of immune homeostasis. gdTreg cells may contribute to the tolerogenic effect of POIT within the first 24-weeks of POIT treatment. These findings suggest that gdTregs cells may be an early marker of desensitization in subjects undergoing POIT.


Assuntos
Arachis/imunologia , Dessensibilização Imunológica/métodos , Genes Codificadores da Cadeia delta de Receptores de Linfócitos T , Genes Codificadores da Cadeia gama de Receptores de Linfócitos T , Hipersensibilidade a Amendoim/terapia , Linfócitos T Reguladores/imunologia , Administração Oral , Alérgenos/administração & dosagem , Criança , Pré-Escolar , Humanos , Memória Imunológica , Família Multigênica , Hipersensibilidade a Amendoim/genética , Hipersensibilidade a Amendoim/imunologia , RNA-Seq , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Análise de Célula Única , Fatores de Tempo , Transcriptoma
7.
J Allergy Clin Immunol ; 145(4): 1219-1230, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31838046

RESUMO

BACKGROUND: Unexpected allergic reactions to peanut are the most common cause of fatal food-related anaphylaxis. Mechanisms underlying the variable severity of peanut-allergic reactions remain unclear. OBJECTIVES: We sought to expand mechanistic understanding of reaction severity in peanut allergy. METHODS: We performed an integrated transcriptomic and epigenomic study of peanut-allergic children as they reacted in vivo during double-blind, placebo-controlled peanut challenges. We integrated whole-blood transcriptome and CD4+ T-cell epigenome profiles to identify molecular signatures of reaction severity (ie, how severely a peanut-allergic child reacts when exposed to peanut). A threshold-weighted reaction severity score was calculated for each subject based on symptoms experienced during peanut challenge and the eliciting dose. Through linear mixed effects modeling, network construction, and causal mediation analysis, we identified genes, CpGs, and their interactions that mediate reaction severity. Findings were replicated in an independent cohort. RESULTS: We identified 318 genes with changes in expression during the course of reaction associated with reaction severity, and 203 CpG sites with differential DNA methylation associated with reaction severity. After replicating these findings in an independent cohort, we constructed interaction networks with the identified peanut severity genes and CpGs. These analyses and leukocyte deconvolution highlighted neutrophil-mediated immunity. We identified NFKBIA and ARG1 as hubs in the networks and 3 groups of interacting key node CpGs and peanut severity genes encompassing immune response, chemotaxis, and regulation of macroautophagy. In addition, we found that gene expression of PHACTR1 and ZNF121 causally mediates the association between methylation at corresponding CpGs and reaction severity, suggesting that methylation may serve as an anchor upon which gene expression modulates reaction severity. CONCLUSIONS: Our findings enhance current mechanistic understanding of the genetic and epigenetic architecture of reaction severity in peanut allergy.


Assuntos
Anafilaxia/genética , Linfócitos T CD4-Positivos/fisiologia , Hipersensibilidade a Amendoim/genética , Adolescente , Alérgenos/imunologia , Arachis/imunologia , Criança , Estudos de Coortes , Metilação de DNA , Progressão da Doença , Epigênese Genética , Feminino , Redes Reguladoras de Genes , Humanos , Imunidade/genética , Imunização , Masculino , Transcriptoma
8.
Clin Immunol ; 219: 108568, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32783912

RESUMO

Oral immunotherapy (OIT) can successfully desensitize allergic individuals to offending foods such as peanut. Our recent clinical trial (NCT02103270) of peanut OIT allowed us to monitor peanut-specific CD4+ T cells, using MHC-peptide Dextramers, over the course of OIT. We used a single-cell targeted RNAseq assay to analyze these cells at 0, 12, 24, 52, and 104 weeks of OIT. We found a transient increase in TGFß-producing cells at 52 weeks in those with successful desensitization, which lasted until 117 weeks. We also performed clustering and identified 5 major clusters of Dextramer+ cells, which we tracked over time. One of these clusters appeared to be anergic, while another was consistent with recently described TFH13 cells. The other 3 clusters appeared to be Th2 cells by their coordinated production of IL-4 and IL-13, but they varied in their expression of STAT signaling proteins and other markers. A cluster with high expression of STAT family members also showed a possible transient increase at week 24 in those with successful desensitization. Single cell TCRαß repertoire sequences were too diverse to track clones over time. Together with increased TGFß production, these changes may be mechanistic predictors of successful OIT that should be further investigated.


Assuntos
Alérgenos/imunologia , Arachis/imunologia , Linfócitos T CD4-Positivos/imunologia , Dessensibilização Imunológica , Hipersensibilidade a Amendoim , Administração Oral , Adolescente , Adulto , Criança , Método Duplo-Cego , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Hipersensibilidade a Amendoim/genética , Hipersensibilidade a Amendoim/imunologia , Hipersensibilidade a Amendoim/terapia , RNA-Seq , Transcrição Gênica , Fator de Crescimento Transformador beta1/genética , Adulto Jovem
9.
J Allergy Clin Immunol ; 143(3): 1027-1037.e7, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30342892

RESUMO

BACKGROUND: Improved animal models are needed to understand the genetic and environmental factors that contribute to food allergy. OBJECTIVE: We sought to assess food allergy phenotypes in a genetically diverse collection of mice. METHODS: We selected 16 Collaborative Cross (CC) mouse strains, as well as the classic inbred C57BL/6J, C3H/HeJ, and BALB/cJ strains, for screening. Female mice were sensitized to peanut intragastrically with or without cholera toxin and then challenged with peanut by means of oral gavage or intraperitoneal injection and assessed for anaphylaxis. Peanut-specific immunoglobulins, T-cell cytokines, regulatory T cells, mast cells, and basophils were quantified. RESULTS: Eleven of the 16 CC strains had allergic reactions to intraperitoneal peanut challenge, whereas only CC027/GeniUnc mice reproducibly experienced severe symptoms after oral food challenge (OFC). CC027/GeniUnc, C3H/HeJ, and C57BL/6J mice all mounted a TH2 response against peanut, leading to production of IL-4 and IgE, but only the CC027/GeniUnc mice reacted to OFC. Orally induced anaphylaxis in CC027/GeniUnc mice was correlated with serum levels of Ara h 2 in circulation but not with allergen-specific IgE or mucosal mast cell protease 1 levels, indicating systemic allergen absorption is important for anaphylaxis through the gastrointestinal tract. Furthermore, CC027/GeniUnc, but not C3H/HeJ or BALB/cJ, mice can be sensitized in the absence of cholera toxin and react on OFC to peanut. CONCLUSIONS: We have identified and characterized CC027/GeniUnc mice as a strain that is genetically susceptible to peanut allergy and prone to severe reactions after OFC. More broadly, these findings demonstrate the untapped potential of the CC population in developing novel models for allergy research.


Assuntos
Alérgenos/efeitos adversos , Arachis/efeitos adversos , Hipersensibilidade a Amendoim/genética , Alérgenos/imunologia , Animais , Arachis/imunologia , Toxina da Cólera/administração & dosagem , Citocinas/genética , Feminino , Variação Genética , Imunoglobulina E/imunologia , Jejuno/imunologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Hipersensibilidade a Amendoim/imunologia , Especificidade da Espécie , Baço/imunologia
10.
Immunology ; 157(2): 110-121, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30779114

RESUMO

Baseline eosinophils reside in the gastrointestinal tract; however, in several allergic disorders, excessive eosinophils accumulate in the blood as well in the tissues. Recently, we showed in vitro that interleukin (IL)-18 matures and transforms IL-5-generated eosinophils into the pathogenic eosinophils that are detected in human allergic diseases. To examine the role of local induction of IL-18 in promoting eosinophil-associated intestinal disorders, we generated enterocyte IL-18-overexpressing mice using the rat intestinal fatty acid-binding promoter (Fabpi) and analysed tissue IL-18 overexpression and eosinophilia by performing real-time polymerase chain reaction, Enzyme-Linked Immunosorbent Assay and anti-major basic protein immunostaining. Herein we show that Fabpi-IL-18 mice display highly induced IL-18 mRNA and protein in the jejunum. IL-18 overexpression in enterocytes promotes marked increases of eosinophils in the blood and jejunum. Our analysis shows IL-18 overexpression in the jejunum induces a specific population of CD101+  CD274+ tissue eosinophils. Additionally, we observed comparable tissue eosinophilia in IL-13-deficient-Fabpi-IL-18 mice, and reduced numbers of tissue eosinophils in eotaxin-deficient-Fabpi-IL-18 and IL-5-deficient-Fabpi-IL-18 mice compared with Fabpi-IL-18 transgenic mice. Notably, jejunum eosinophilia in IL-5-deficient-Fabpi-IL-18 mice is significantly induced compared with wild-type mice, which indicates the direct role of induced IL-18 in the tissue accumulation of eosinophils and mast cells. Furthermore, we also found that overexpression of IL-18 in the intestine promotes eosinophil-associated peanut-induced allergic responses in mice. Taken together, we provide direct in vivo evidence that induced expression of IL-18 in the enterocytes promotes eotaxin-1, IL-5 and IL-13 independent intestinal eosinophilia, which signifies the clinical relevance of induced IL-18 in eosinophil-associated gastrointestinal disorders (EGIDs) to food allergens.


Assuntos
Enterócitos/imunologia , Eosinófilos/imunologia , Interleucina-18/imunologia , Jejuno/imunologia , Hipersensibilidade a Amendoim/imunologia , Animais , Antígenos CD/genética , Antígenos CD/imunologia , Antígeno B7-H1/genética , Antígeno B7-H1/imunologia , Quimiocina CCL11/genética , Quimiocina CCL11/imunologia , Enterócitos/patologia , Eosinófilos/patologia , Humanos , Interleucina-13/genética , Interleucina-13/imunologia , Interleucina-18/genética , Interleucina-5/genética , Interleucina-5/imunologia , Jejuno/patologia , Mastócitos/imunologia , Mastócitos/patologia , Camundongos , Camundongos Transgênicos , Hipersensibilidade a Amendoim/genética , Hipersensibilidade a Amendoim/patologia , Ratos
11.
J Immunol ; 199(9): 3116-3128, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28972089

RESUMO

CD22, a sialic acid-binding Ig-type lectin (Siglec) family member, is an inhibitory coreceptor of the BCR with established roles in health and disease. The restricted expression pattern of CD22 on B cells and most B cell lymphomas has made CD22 a therapeutic target for B cell-mediated diseases. Models to better understand how in vivo targeting of CD22 translates to human disease are needed. In this article, we report the development of a transgenic mouse expressing human CD22 (hCD22) in B cells and assess its ability to functionally substitute for murine CD22 (mCD22) for regulation of BCR signaling, Ab responses, homing, and tolerance. Expression of hCD22 on transgenic murine B cells is comparable to expression on human primary B cells, and it colocalizes with mCD22 on the cell surface. Murine B cells expressing only hCD22 have identical calcium (Ca2+) flux responses to anti-IgM as mCD22-expressing wild-type B cells. Furthermore, hCD22 transgenic mice on an mCD22-/- background have restored levels of marginal zone B cells and Ab responses compared with deficiencies observed in CD22-/- mice. Consistent with these observations, hCD22 transgenic mice develop normal humoral responses in a peanut allergy oral sensitization model. Homing of B cells to Peyer's patches was partially rescued by expression of hCD22 compared with CD22-/- B cells, although not to wild-type levels. Notably, Siglec-engaging antigenic liposomes formulated with an hCD22 ligand were shown to prevent B cell activation, increase cell death, and induce tolerance in vivo. This hCD22 transgenic mouse will be a valuable model for investigating the function of hCD22 and preclinical studies targeting hCD22.


Assuntos
Linfócitos B/imunologia , Hipersensibilidade a Amendoim/imunologia , Nódulos Linfáticos Agregados/imunologia , Receptores de Antígenos de Linfócitos B/imunologia , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia , Transdução de Sinais/imunologia , Animais , Linfócitos B/patologia , Modelos Animais de Doenças , Humanos , Ativação Linfocitária/genética , Camundongos , Camundongos Transgênicos , Hipersensibilidade a Amendoim/genética , Hipersensibilidade a Amendoim/patologia , Nódulos Linfáticos Agregados/patologia , Receptores de Antígenos de Linfócitos B/genética , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico/genética , Transdução de Sinais/genética
12.
J Allergy Clin Immunol ; 141(3): 991-1001, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29030101

RESUMO

BACKGROUND: Peanut allergy (PA) is a complex disease with both environmental and genetic risk factors. Previously, PA loci were identified in filaggrin (FLG) and HLA in candidate gene studies, and loci in HLA were identified in a genome-wide association study and meta-analysis. OBJECTIVE: We sought to investigate genetic susceptibility to PA. METHODS: Eight hundred fifty cases and 926 hyper-control subjects and more than 7.8 million genotyped and imputed single nucleotide polymorphisms (SNPs) were analyzed in a genome-wide association study to identify susceptibility variants for PA in the Canadian population. A meta-analysis of 2 phenotypes (PA and food allergy) was conducted by using 7 studies from the Canadian, American (n = 2), Australian, German, and Dutch (n = 2) populations. RESULTS: An SNP near integrin α6 (ITGA6) reached genome-wide significance with PA (P = 1.80 × 10-8), whereas SNPs associated with Src kinase-associated phosphoprotein 1 (SKAP1), matrix metallopeptidase 12 (MMP12)/MMP13, catenin α3 (CTNNA3), rho GTPase-activating protein 24 (ARHGAP24), angiopoietin 4 (ANGPT4), chromosome 11 open reading frame (C11orf30/EMSY), and exocyst complex component 4 (EXOC4) reached a threshold suggestive of association (P ≤ 1.49 × 10-6). In the meta-analysis of PA, loci in or near ITGA6, ANGPT4, MMP12/MMP13, C11orf30, and EXOC4 were significant (P ≤ 1.49 × 10-6). When a phenotype of any food allergy was used for meta-analysis, the C11orf30 locus reached genome-wide significance (P = 7.50 × 10-11), whereas SNPs associated with ITGA6, ANGPT4, MMP12/MMP13, and EXOC4 and additional C11orf30 SNPs were suggestive (P ≤ 1.49 × 10-6). Functional annotation indicated that SKAP1 regulates expression of CBX1, which colocalizes with the EMSY protein coded by C11orf30. CONCLUSION: This study identifies multiple novel loci as risk factors for PA and food allergy and establishes C11orf30 as a risk locus for both PA and food allergy. Multiple genes (C11orf30/EMSY, SKAP1, and CTNNA3) identified by this study are involved in epigenetic regulation of gene expression.


Assuntos
Epigênese Genética , Loci Gênicos , Estudo de Associação Genômica Ampla , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Hipersensibilidade a Amendoim/genética , Polimorfismo de Nucleotídeo Único , Proteínas Repressoras/genética , Homólogo 5 da Proteína Cromobox , Feminino , Proteínas Filagrinas , Humanos , Masculino , Hipersensibilidade a Amendoim/epidemiologia , Hipersensibilidade a Amendoim/metabolismo , Fosfoproteínas/biossíntese , Fosfoproteínas/genética , Fatores de Risco , alfa Catenina/biossíntese , alfa Catenina/genética
13.
J Hum Genet ; 63(2): 239-248, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29192240

RESUMO

B-cell receptors (BCRs) play a critical role in adaptive immunity as they generate highly diverse immunoglobulin repertoires to recognize a wide variety of antigens. To better understand immune responses, it is critically important to establish a quantitative and rapid method to analyze BCR repertoire comprehensively. Here, we developed "Bcrip", a novel approach to characterize BCR repertoire by sequencing millions of BCR cDNA using next-generation sequencer. Using this method and quantitative real-time PCR, we analyzed expression levels and repertoires of BCRs in a total of 17 peanut allergic subjects' peripheral blood samples before and after receiving oral immunotherapy (OIT) or placebo. By our methods, we successfully identified all of variable (V), joining (J), and constant (C) regions, in an average of 79.1% of total reads and 99.6% of these VJC-mapped reads contained the C region corresponding to the isotypes that we aimed to analyze. In the 17 peanut allergic subjects' peripheral blood samples, we observed an oligoclonal enrichment of certain immunoglobulin heavy chain alpha (IGHA) and IGH gamma (IGHG) clones (P = 0.034 and P = 0.027, respectively) in peanut allergic subjects after OIT. This newly developed BCR sequencing and analysis method can be applied to investigate B-cell repertoires in various research areas, including food allergies as well as autoimmune and infectious diseases.


Assuntos
Imunoterapia , Hipersensibilidade a Amendoim/genética , Hipersensibilidade a Amendoim/terapia , Receptores de Antígenos de Linfócitos B/genética , Criança , Pré-Escolar , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Hipersensibilidade a Amendoim/imunologia , Receptores de Antígenos de Linfócitos B/imunologia
14.
Trans Am Clin Climatol Assoc ; 129: 250-265, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30166721

RESUMO

Advances in understanding the immunological basis and mechanisms underlying allergic and immunologic disorders have led to effective but costly long-term and repetitive biologic therapies. Gene therapy is a rapidly advancing technology, in which a single administration of an adeno-associated virus encoding the therapeutic protein or monoclonal antibody may provide effective long-term therapy for allergic and immunologic disorders. In this review, we summarize the recent studies from our laboratory developing gene therapy strategies to treat hereditary angioedema and peanut allergy. The unraveling of the pathogenesis of immune-based disorders, including hereditary deficiencies of components of the immune system and allergic disorders, has led to the development of therapies using parenteral administration of recombinant proteins or monoclonal antibodies (1). While many of these therapies are highly effective, they are limited by the half-life of the therapeutic protein or antibody, requiring repetitive administration of days to weeks (2-15). The focus of recent work in our laboratory has been to solve this problem by substituting protein/monoclonal antibody administration with gene therapy, where current technology allows for a single administration of the gene coding for a protein or antibody to provide persistent expression of effective levels of the therapeutic protein or antibody. Gene therapy is a drug delivery platform which uses genetic material, usually in the form of coding exons of the therapeutic gene, to correct, compensate for, or prevent the development of an abnormal phenotype (16). Originally conceptualized as a strategy to treat rare hereditary disorders, gene therapy is being developed for a wide range of human disorders, including common acquired conditions (17-20). In this review, we will describe how we have adopted gene therapy technology to develop therapies for immune-related disorders, using as examples hereditary angioedema, an inherited autosomal dominant disorder, and peanut allergy, a common acquired allergic disorder.


Assuntos
Angioedemas Hereditários/terapia , Terapia Genética/métodos , Imunoterapia/métodos , Hipersensibilidade a Amendoim/terapia , Angioedemas Hereditários/genética , Angioedemas Hereditários/imunologia , Animais , Humanos , Hipersensibilidade a Amendoim/genética , Hipersensibilidade a Amendoim/imunologia , Resultado do Tratamento
15.
Clin Exp Allergy ; 47(2): 217-223, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27883235

RESUMO

BACKGROUND: Genetic variants for IgE-mediated peanut allergy are yet to be fully characterized and to date only one genomewide association study (GWAS) has been published. OBJECTIVE: To identify genetic variants associated with challenge-proven peanut allergy. METHODS: We carried out a GWAS comparing 73 infants with challenge-proven IgE-mediated peanut allergy against 148 non-allergic infants (all ~ 1 year old). We tested a total of 3.8 million single nucleotide polymorphisms, as well as imputed HLA alleles and amino acids. Replication was assessed by de novo genotyping in a panel of additional 117 cases and 380 controls, and in silico testing in two independent GWAS cohorts. RESULTS: We identified 21 independent associations at P ≤ 5 × 10-5 but were unable to replicate these. The most significant HLA association was the previously reported amino acid variant located at position 71, within the peptide-binding groove of HLA-DRB1 (P = 2 × 10-4 ). Our study therefore reproduced previous findings for the association between peanut allergy and HLA-DRB1 in this Australian population. CONCLUSIONS AND CLINICAL RELEVANCE: Genetic determinants for challenge-proven peanut allergy include alleles at the HLA-DRB1 locus.


Assuntos
Substituição de Aminoácidos , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Cadeias HLA-DRB1/genética , Hipersensibilidade a Amendoim/genética , Hipersensibilidade a Amendoim/imunologia , Polimorfismo Genético , Alelos , Genótipo , Cadeias HLA-DRB1/química , Cadeias HLA-DRB1/imunologia , Humanos , Razão de Chances , Polimorfismo de Nucleotídeo Único
16.
Clin Exp Allergy ; 47(10): 1331-1341, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28802077

RESUMO

BACKGROUND: Food allergy is an increasingly common health problem in Western populations. Epidemiological studies have suggested both positive and negative associations between food allergy and infection with the gastric bacterium Helicobacter pylori. OBJECTIVE: The objective of this work was to investigate whether experimental infection with H. pylori, or prophylactic treatment with H. pylori-derived immunomodulatory molecules, affects the onset and severity of food allergy, either positively or negatively. METHODS: We infected neonatal C57BL/6 or C3H mice with H. pylori or treated animals with H. pylori components (bacterial lysate or the immunomodulator VacA) and subsequently subjected them to four different protocols for food allergy induction, using either ovalbumin or peanut extract as allergens for sensitization and challenge. Readouts included anaphylaxis scoring, quantification of allergen-specific serum IgE and IgG1 and of the mast cell protease MCPT1, as well as splenic T-helper-2 cell-derived cytokine production. Mesenteric lymph node CD4+ FoxP3+ regulatory T cells were subjected to flow cytometric quantification and sorting followed by qRT-PCR, and to DNA methylation analyses of the Treg-specific demethylated region (TSDR) within the FOXP3 locus. RESULTS: Mice that had been infected with H. pylori or treated with H. pylori-derived immunomodulators showed reduced anaphylaxis upon allergen sensitization and challenge, irrespective of the allergen used. Most of the immunologic assays confirmed a protective effect of H. pylori. CD4+ FoxP3+ T cells were more abundant in protected mice and exhibited a stable Treg phenotype characterized by FOXP3 TSDR demethylation. CONCLUSIONS AND CLINICAL RELEVANCE: Helicobacter pylori confers protection against the anaphylaxis associated with ovalbumin and peanut allergy and affects the epigenome of T cells, thereby promoting stable Treg differentiation and functionality. Prophylactic treatment with H. pylori-derived immunomodulators appears to be a promising strategy for food allergy prevention.


Assuntos
Anafilaxia/prevenção & controle , Proteínas de Bactérias/imunologia , Hipersensibilidade Alimentar/prevenção & controle , Helicobacter pylori/imunologia , Fatores Imunológicos/imunologia , Alérgenos/imunologia , Anafilaxia/genética , Anafilaxia/imunologia , Anafilaxia/metabolismo , Animais , Ilhas de CpG , Citocinas/sangue , Citocinas/metabolismo , Metilação de DNA , Modelos Animais de Doenças , Hipersensibilidade Alimentar/genética , Hipersensibilidade Alimentar/imunologia , Hipersensibilidade Alimentar/metabolismo , Imunoglobulina E/imunologia , Masculino , Camundongos , Hipersensibilidade a Amendoim/genética , Hipersensibilidade a Amendoim/imunologia , Hipersensibilidade a Amendoim/metabolismo , Hipersensibilidade a Amendoim/prevenção & controle , Baço/citologia , Baço/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo
17.
Allergy ; 72(7): 1081-1090, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28106273

RESUMO

BACKGROUND: The pathology of allergic diseases involves type 2 immune cells, such as Th2, ILC2, and basophils exerting their effect by production of IL-4, IL-5, and IL-13. However, surface receptors that are specifically expressed on type 2 immune cells are less well documented. The aim of this investigation was to identify surface markers associated with type 2 inflammation. METHODS: Naïve human CD4+ T cells were short-term activated in the presence or absence of IL-4 and analyzed for expression of >300 cell-surface proteins. Ex vivo-isolated peripheral blood mononuclear cells (PBMCs) from peanut-allergic (PA) and nonallergic subjects were stimulated (14-16 h) with peanut extract to detect peanut-specific CD4+ CD154+ T cells. Biopsies were obtained for transcriptomic analysis from healthy controls and patients with extrinsic or intrinsic atopic dermatitis (AD) and psoriasis. RESULTS: Expression analysis of >300 surface proteins enabled identification of IL-4-upregulated surface proteins, such as CD90, CD108, CD109, and CD200R (CD200R1). Additional analysis of in vitro-differentiated Th0, Th1, and Th2 cultures identified CD200R as upregulated on Th2 cells. From ex vivo-isolated PBMCs, we found high expression of CD200R on Th2 and ILC2 cells and basophils. In PA subjects, the peanut-specific Th2 (CD154+ CRTh2+ ) cells expressed more CD200R than the non-allergen-specific Th2 (CD154- CRTh2+ ) cells. Moreover, costaining of CD161 and CD200R identified peanut-specific highly differentiated IL-4+ IL-5+ Th2 cells. Finally, transcriptomic analysis revealed upregulation of CD200R in lesional skin from subjects with an extrinsic AD phenotype compared to healthy skin. CONCLUSION: These results indicate that CD200R expression strongly correlates with Th2 pathology; though, the mechanism is as yet elusive.


Assuntos
Antígenos de Superfície/genética , Receptores de Superfície Celular/genética , Células Th2/imunologia , Células Th2/metabolismo , Alérgenos/imunologia , Antígenos de Superfície/metabolismo , Basófilos/imunologia , Basófilos/metabolismo , Citocinas/metabolismo , Expressão Gênica , Humanos , Hipersensibilidade/genética , Hipersensibilidade/imunologia , Hipersensibilidade/metabolismo , Receptores de Orexina , Hipersensibilidade a Amendoim/genética , Hipersensibilidade a Amendoim/imunologia , Hipersensibilidade a Amendoim/metabolismo , Receptores de Superfície Celular/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Antígenos Thy-1/metabolismo
20.
J Allergy Clin Immunol ; 135(5): 1283-92, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25542883

RESUMO

BACKGROUND: Studies suggest that oral immunotherapy (OIT) and sublingual immunotherapy (SLIT) for food allergy hold promise; however, the immunologic mechanisms underlying these therapies are not well understood. OBJECTIVE: We sought to generate insights into the mechanisms and duration of suppression of immune responses to peanut during immunotherapy. METHODS: Blood was obtained from subjects at baseline and at multiple time points during a placebo-controlled trial of peanut OIT and SLIT. Immunologic outcomes included measurement of spontaneous and stimulated basophil activity by using automated fluorometry (histamine) and flow cytometry (activation markers and IL-4), measurement of allergen-induced cytokine expression in dendritic cell (DC)-T-cell cocultures by using multiplexing technology, and measurement of MHC II and costimulatory molecule expression on DCs by using flow cytometry. RESULTS: Spontaneous and allergen-induced basophil reactivity (histamine release, CD63 expression, and IL-4 production) were suppressed during dose escalation and after 6 months of maintenance dosing. Peanut- and dust mite-induced expression of TH2 cytokines was reduced in DC-T-cell cocultures during immunotherapy. This was associated with decreased levels of CD40, HLA-DR, and CD86 expression on DCs and increased expression of CD80. These effects were most striking in myeloid DC-T-cell cocultures from subjects receiving OIT. Many markers of immunologic suppression reversed after withdrawal from immunotherapy and in some cases during ongoing maintenance therapy. CONCLUSION: OIT and SLIT for peanut allergy induce rapid suppression of basophil effector functions, DC activation, and TH2 cytokine responses during the initial phases of immunotherapy in an antigen-nonspecific manner. Although there was some interindividual variation, in many patients suppression appeared to be temporary.


Assuntos
Dessensibilização Imunológica , Hipersensibilidade a Amendoim/imunologia , Hipersensibilidade a Amendoim/terapia , Administração Oral , Administração Sublingual , Alérgenos/administração & dosagem , Alérgenos/imunologia , Arachis/efeitos adversos , Basófilos/imunologia , Basófilos/metabolismo , Biomarcadores , Citocinas/metabolismo , Células Dendríticas/imunologia , Expressão Gênica , Antígenos HLA-DR/genética , Antígenos HLA-DR/imunologia , Humanos , Interleucina-4/metabolismo , Hipersensibilidade a Amendoim/genética , Projetos Piloto , Linfócitos T/imunologia , Linfócitos T/metabolismo , Tetraspanina 30/metabolismo , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA