1
|
Arnau‐Soler A, Tremblay BL, Sun Y, Madore A, Simard M, Kersten ETG, Ghauri A, Marenholz I, Eiwegger T, Simons E, Chan ES, Nadeau K, Sampath V, Mazer BD, Elliott S, Hampson C, Soller L, Sandford A, Begin P, Hui J, Wilken BF, Gerdts J, Bourkas A, Ellis AK, Vasileva D, Clarke A, Eslami A, Ben‐Shoshan M, Martino D, Daley D, Koppelman GH, Laprise C, Lee Y, Asai Y. Food Allergy Genetics and Epigenetics: A Review of Genome-Wide Association Studies. Allergy 2025; 80:106-131. [PMID: 39698764 PMCID: PMC11724255 DOI: 10.1111/all.16429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 10/12/2024] [Accepted: 11/26/2024] [Indexed: 12/20/2024]
Abstract
In this review, we provide an overview of food allergy genetics and epigenetics aimed at clinicians and researchers. This includes a brief review of the current understanding of genetic and epigenetic mechanisms, inheritance of food allergy, as well as a discussion of advantages and limitations of the different types of studies in genetic research. We specifically focus on the results of genome-wide association studies in food allergy, which have identified 16 genetic variants that reach genome-wide significance, many of which overlap with other allergic diseases, including asthma, atopic dermatitis, and allergic rhinitis. Identified genes for food allergy are mainly involved in epithelial barrier function (e.g., FLG, SERPINB7) and immune function (e.g., HLA, IL4). Epigenome-wide significant findings at 32 loci are also summarized as well as 14 additional loci with significance at a false discovery of < 1 × 10-4. Integration of epigenetic and genetic data is discussed in the context of disease mechanisms, many of which are shared with other allergic diseases. The potential utility of genetic and epigenetic discoveries is deliberated. In the future, genetic and epigenetic markers may offer ways to predict the presence or absence of clinical IgE-mediated food allergy among sensitized individuals, likelihood of development of natural tolerance, and response to immunotherapy.
Collapse
Affiliation(s)
- Aleix Arnau‐Soler
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC)BerlinGermany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität Zu BerlinBerlinGermany
- Experimental and Clinical Research Center, a Joint Cooperation of Max Delbruck Center for Molecular Medicine and Charité—Universitätsmedizin BerlinBerlinGermany
- German Center for Child and Adolescent Health (DZKJ)BerlinGermany
| | - Bénédicte L. Tremblay
- Département Des Sciences FondamentalesUniversité du Québec à ChicoutimiSaguenayQuebecCanada
| | - Yidan Sun
- Department of Pediatric Pulmonology and Pediatric AllergologyUniversity Medical Center Groningen, Beatrix Children's Hospital, University of GroningenGroningenthe Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)Groningenthe Netherlands
| | - Anne‐Marie Madore
- Département Des Sciences FondamentalesUniversité du Québec à ChicoutimiSaguenayQuebecCanada
| | - Mathieu Simard
- Département Des Sciences FondamentalesUniversité du Québec à ChicoutimiSaguenayQuebecCanada
| | - Elin T. G. Kersten
- Department of Pediatric Pulmonology and Pediatric AllergologyUniversity Medical Center Groningen, Beatrix Children's Hospital, University of GroningenGroningenthe Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)Groningenthe Netherlands
| | - Ahla Ghauri
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC)BerlinGermany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität Zu BerlinBerlinGermany
- Experimental and Clinical Research Center, a Joint Cooperation of Max Delbruck Center for Molecular Medicine and Charité—Universitätsmedizin BerlinBerlinGermany
- German Center for Child and Adolescent Health (DZKJ)BerlinGermany
| | - Ingo Marenholz
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC)BerlinGermany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität Zu BerlinBerlinGermany
- Experimental and Clinical Research Center, a Joint Cooperation of Max Delbruck Center for Molecular Medicine and Charité—Universitätsmedizin BerlinBerlinGermany
| | - Thomas Eiwegger
- Translational Medicine Program, Research InstituteHospital for Sick ChildrenTorontoOntarioCanada
- Department of Immunology, Temerty Faculty of MedicineUniversity of TorontoTorontoOntarioCanada
- Karl Landsteiner University of Health SciencesKrems an der DonauAustria
- Department of Pediatric and Adolescent MedicineUniversity Hospital St. PöltenSt. PöltenAustria
- Department of Paediatrics, Division of Clinical Immunology and Allergy, Food Allergy and Anaphylaxis Program, the Hospital for Sick ChildrenThe University of TorontoTorontoOntarioCanada
| | - Elinor Simons
- Section of Allergy & Clinical Immunology, Department of Pediatrics & Child Health, University of ManitobaChildren's Hospital Research InstituteWinnipegManitobaCanada
| | - Edmond S. Chan
- Division of Allergy, Department of PediatricsThe University of British ColumbiaVancouverBritish ColumbiaCanada
| | - Kari Nadeau
- Department of Environmental StudiesHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | - Vanitha Sampath
- Department of Environmental StudiesHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | - Bruce D. Mazer
- Research Institute of the McGill University Health CentreMontrealQuebecCanada
| | - Susan Elliott
- Department of Geography and Environmental ManagementUniversity of WaterlooWaterlooOntarioCanada
| | | | - Lianne Soller
- Division of Allergy, Department of PediatricsThe University of British ColumbiaVancouverBritish ColumbiaCanada
| | - Andrew Sandford
- Department of MedicineThe University of British ColumbiaVancouverBritish ColumbiaCanada
- Centre for Heart Lung InnovationVancouverBritish ColumbiaCanada
| | - Philippe Begin
- Department of Pediatrics, Service of Allergy and Clinical ImmunologyCentre Hospitalier Universitaire Sainte‐JustineMontréalQuébecCanada
- Department of Medicine, Service of Allergy and Clinical ImmunologyCentre Hospitalier de l'Université de MontréalMontréalQuébecCanada
| | - Jennie Hui
- School of Population HealthUniversity of Western AustraliaPerthWestern AustraliaAustralia
| | - Bethany F. Wilken
- School of Medicine, Department of MedicineQueen's UniversityKingstonOntarioCanada
| | | | - Adrienn Bourkas
- School of Medicine, Department of MedicineQueen's UniversityKingstonOntarioCanada
| | - Anne K. Ellis
- Division of Allergy & Immunology, Department of MedicineQueen's UniversityKingstonOntarioCanada
| | - Denitsa Vasileva
- Department of MedicineThe University of British ColumbiaVancouverBritish ColumbiaCanada
- Centre for Heart Lung InnovationVancouverBritish ColumbiaCanada
| | - Ann Clarke
- Department of Medicine, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
| | - Aida Eslami
- Département de médecine Sociale et préventive, Faculté de médecineUniversité LavalQuebecCanada
| | - Moshe Ben‐Shoshan
- Division of Allergy and Clinical Immunology, Department of Pediatrics, Montréal Children's HospitalMcGill University Health CentreMontréalQuebecCanada
| | - David Martino
- Wal‐Yan Respiratory Research CentreTelethon Kids InstitutePerthAustralia
| | - Denise Daley
- Department of MedicineThe University of British ColumbiaVancouverBritish ColumbiaCanada
- Centre for Heart Lung InnovationVancouverBritish ColumbiaCanada
| | - Gerard H. Koppelman
- Department of Pediatric Pulmonology and Pediatric AllergologyUniversity Medical Center Groningen, Beatrix Children's Hospital, University of GroningenGroningenthe Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)Groningenthe Netherlands
| | - Catherine Laprise
- Département Des Sciences FondamentalesUniversité du Québec à ChicoutimiSaguenayQuebecCanada
| | - Young‐Ae Lee
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC)BerlinGermany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität Zu BerlinBerlinGermany
- Experimental and Clinical Research Center, a Joint Cooperation of Max Delbruck Center for Molecular Medicine and Charité—Universitätsmedizin BerlinBerlinGermany
- German Center for Child and Adolescent Health (DZKJ)BerlinGermany
| | - Yuka Asai
- Division of Dermatology, Department of MedicineQueen's UniversityKingstonOntarioCanada
| |
Collapse
|
2
|
Imran S, Neeland MR, Martino DJ, Peng S, Koplin J, Dharmage SC, Tang MLK, Sawyer S, Dang T, McWilliam V, Peters RL, Prescott S, Perrett KP, Novakovic B, Saffery R. Epigenomic variability is associated with age-specific naïve CD4 T cell response to activation in infants and adolescents. Immunol Cell Biol 2023; 101:397-411. [PMID: 36760028 PMCID: PMC10952707 DOI: 10.1111/imcb.12628] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/11/2023]
Abstract
Childhood is a critical period of immune development. During this time, naïve CD4 (nCD4) T cells undergo programmed cell differentiation, mediated by epigenetic changes, in response to external stimuli leading to a baseline homeostatic state that may determine lifelong disease risk. However, the ontogeny of epigenetic signatures associated with CD4 T cell activation during key developmental periods are yet to be described. We investigated genome-wide DNA methylation (DNAm) changes associated with nCD4 T activation following 72 h culture in media+anti-CD3/CD28 beads in healthy infants (aged 12 months, n = 18) and adolescents (aged 10-15 years, n = 15). We integrated these data with transcriptomic and cytokine profiling from the same samples. nCD4 T cells from both age groups show similar extensive epigenetic reprogramming following activation, with the majority of genes involved in the T cell receptor signaling pathway associated with differential methylation. Additionally, we identified differentially methylated probes showing age-specific responses, that is, responses in only infants or adolescents, including within a cluster of T cell receptor (TCR) genes. These encoded several TCR alpha joining (TRAJ), and TCR alpha variable (TRAV) genes. Cytokine data analysis following stimulation revealed enhanced release of IFN-γ, IL-2 and IL-10, in nCD4 T cells from adolescents compared with infants. Overlapping differential methylation and cytokine responses identified four probes potentially underpinning these age-specific responses. We show that DNAm in nCD4T cells in response to activation is dynamic in infancy and adolescence, with additional evidence for age-specific effects potentially driving variation in cytokine responses between these ages.
Collapse
Affiliation(s)
- Samira Imran
- Murdoch Children's Research Institute, and Department of PaediatricsUniversity of Melbourne, Royal Children's HospitalFlemington RoadParkvilleVICAustralia
| | - Melanie R Neeland
- Murdoch Children's Research Institute, and Department of PaediatricsUniversity of Melbourne, Royal Children's HospitalFlemington RoadParkvilleVICAustralia
| | - David J. Martino
- Wal‐yan Respiratory Research Centre, Telethon Kids InstitutePerthAustralia
- University of Western AustraliaPerthWAAustralia
| | - Stephen Peng
- Murdoch Children's Research Institute, and Department of PaediatricsUniversity of Melbourne, Royal Children's HospitalFlemington RoadParkvilleVICAustralia
| | - Jennifer Koplin
- Murdoch Children's Research Institute, and Department of PaediatricsUniversity of Melbourne, Royal Children's HospitalFlemington RoadParkvilleVICAustralia
| | - Shyamali C Dharmage
- Murdoch Children's Research Institute, and Department of PaediatricsUniversity of Melbourne, Royal Children's HospitalFlemington RoadParkvilleVICAustralia
- Allergy and Lung Health UnitMelbourne School of Population and Global HealthUniversity of MelbourneMelbourneVICAustralia
| | - Mimi LK Tang
- Murdoch Children's Research Institute, and Department of PaediatricsUniversity of Melbourne, Royal Children's HospitalFlemington RoadParkvilleVICAustralia
- Department of Allergy and ImmunologyRoyal Children's HospitalMelbourneVICAustralia
| | - Susan Sawyer
- Murdoch Children's Research Institute, and Department of PaediatricsUniversity of Melbourne, Royal Children's HospitalFlemington RoadParkvilleVICAustralia
- Centre for Adolescent HealthRoyal Children's Hospital MelbourneMelbourneVICAustralia
| | - Thanh Dang
- Murdoch Children's Research Institute, and Department of PaediatricsUniversity of Melbourne, Royal Children's HospitalFlemington RoadParkvilleVICAustralia
| | - Vicki McWilliam
- Murdoch Children's Research Institute, and Department of PaediatricsUniversity of Melbourne, Royal Children's HospitalFlemington RoadParkvilleVICAustralia
- Department of Allergy and ImmunologyRoyal Children's HospitalMelbourneVICAustralia
| | - Rachel L Peters
- Murdoch Children's Research Institute, and Department of PaediatricsUniversity of Melbourne, Royal Children's HospitalFlemington RoadParkvilleVICAustralia
| | - Susan Prescott
- School of MedicineThe University of Western Australia35 Stirling HighwayCrawleyWAAustralia
- Telethon Kids Institute15 Hospital AvenueNedlandsWAAustralia
- Department of ImmunologyPerth Children's Hospital15 Hospital AvenueNedlandsWAAustralia
| | - Kirsten P Perrett
- Murdoch Children's Research Institute, and Department of PaediatricsUniversity of Melbourne, Royal Children's HospitalFlemington RoadParkvilleVICAustralia
- Department of Allergy and ImmunologyRoyal Children's HospitalMelbourneVICAustralia
| | - Boris Novakovic
- Murdoch Children's Research Institute, and Department of PaediatricsUniversity of Melbourne, Royal Children's HospitalFlemington RoadParkvilleVICAustralia
| | - Richard Saffery
- Murdoch Children's Research Institute, and Department of PaediatricsUniversity of Melbourne, Royal Children's HospitalFlemington RoadParkvilleVICAustralia
| |
Collapse
|
3
|
Devonshire A, Gautam Y, Johansson E, Mersha TB. Multi-omics profiling approach in food allergy. World Allergy Organ J 2023; 16:100777. [PMID: 37214173 PMCID: PMC10199264 DOI: 10.1016/j.waojou.2023.100777] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 04/05/2023] [Accepted: 04/05/2023] [Indexed: 05/24/2023] Open
Abstract
The prevalence of food allergy (FA) among children is increasing, affecting nearly 8% of children, and FA is the most common cause of anaphylaxis and anaphylaxis-related emergency department visits in children. Importantly, FA is a complex, multi-system, multifactorial disease mediated by food-specific immunoglobulin E (IgE) and type 2 immune responses and involving environmental and genetic factors and gene-environment interactions. Early exposure to external and internal environmental factors largely influences the development of immune responses to allergens. Genetic factors and gene-environment interactions have established roles in the FA pathophysiology. To improve diagnosis and identification of FA therapeutic targets, high-throughput omics approaches have emerged and been applied over the past decades to screen for potential FA biomarkers, such as genes, transcripts, proteins, and metabolites. In this article, we provide an overview of the current status of FA omics studies, namely genomic, transcriptomic, epigenomic, proteomic, exposomic, and metabolomic. The current development of multi-omics integration of FA studies is also briefly discussed. As individual omics technologies only provide limited information on the multi-system biological processes of FA, integration of population-based multi-omics data and clinical data may lead to robust biomarker discovery that could translate into advances in disease management and clinical care and ultimately lead to precision medicine approaches.
Collapse
Affiliation(s)
- Ashley Devonshire
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Yadu Gautam
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Elisabet Johansson
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Tesfaye B. Mersha
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
4
|
Kazmi W, Berin MC. Oral tolerance and oral immunotherapy for food allergy: Evidence for common mechanisms? Cell Immunol 2023; 383:104650. [PMID: 36543052 DOI: 10.1016/j.cellimm.2022.104650] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/21/2022] [Accepted: 11/26/2022] [Indexed: 12/05/2022]
Abstract
Food allergies affect up to 10% of the US population, can be life-threatening, and have a significant negative impact on quality of life. Delayed dietary introduction of foods in childhood can hinder the induction of oral tolerance, an active regulatory response to foods that prevents the development of food allergy. Some children outgrow their food allergies naturally, while many others have persistent, lifelong food allergy for which there are few therapeutic options. Oral immunotherapy (OIT) is a therapeutic approach of giving increasing amounts of food to attempt to desensitize the allergic individual. In this review, we focus on the immune mechanisms common to oral tolerance and response to oral immunotherapy, with the objective of determining whether true tolerance can be achieved after food allergy has been established.
Collapse
Affiliation(s)
- Wajiha Kazmi
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - M Cecilia Berin
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
5
|
Issa M, Rivière G, Houdeau E, Adel-Patient K. Perinatal exposure to foodborne inorganic nanoparticles: A role in the susceptibility to food allergy? FRONTIERS IN ALLERGY 2022; 3:1067281. [PMID: 36545344 PMCID: PMC9760876 DOI: 10.3389/falgy.2022.1067281] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022] Open
Abstract
Food allergy (FA) is an inappropriate immune response against dietary antigens. Various environmental factors during perinatal life may alter the establishment of intestinal homeostasis, thereby predisposing individuals to the development of such immune-related diseases. Among these factors, recent studies have emphasized the chronic dietary exposure of the mother to foodborne inorganic nanoparticles (NP) such as nano-sized silicon dioxide (SiO2), titanium dioxide (TiO2) or silver (Ag). Indeed, there is growing evidence that these inorganic agents, used as food additives in various products, as processing aids during food manufacturing or in food contact materials, can cross the placental barrier and reach the developing fetus. Excretion in milk is also suggested, hence continuing to expose the neonate during a critical window of susceptibility. Due to their immunotoxical and biocidal properties, such exposure may disrupt the host-intestinal microbiota's beneficial exchanges and may interfere with intestinal barrier and gut-associated immune system development in fetuses then the neonates. The resulting dysregulated intestinal homeostasis in the infant may significantly impede the induction of oral tolerance, a crucial process of immune unresponsiveness to food antigens. The current review focuses on the possible impacts of perinatal exposure to foodborne NP during pregnancy and early life on the susceptibility to developing FA.
Collapse
Affiliation(s)
- Mohammad Issa
- Département Médicaments et Technologies Pour la Santé (MTS), SPI/Laboratoire d’Immuno-Allergie Alimentaire, Université Paris-Saclay, CEA, INRAE, Gif-sur-Yvette, France
| | - Gilles Rivière
- French Agency for Food, Environmental and Occupational Health & Safety (ANSES, Agence Nationale De Sécurité Sanitaire De l’alimentation, De l’environnement et du Travail), Direction de l’Evaluation des Risques, Maisons-Alfort, France
| | - Eric Houdeau
- Toxalim UMR1331 (Research Centre in Food Toxicology), Toulouse University, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Karine Adel-Patient
- Département Médicaments et Technologies Pour la Santé (MTS), SPI/Laboratoire d’Immuno-Allergie Alimentaire, Université Paris-Saclay, CEA, INRAE, Gif-sur-Yvette, France
| |
Collapse
|
6
|
Imran S, Neeland MR, Peng S, Vlahos A, Martino D, Dharmage SC, Tang MLK, Sawyer S, Dang TD, McWilliam V, Peters RL, Koplin JJ, Perrett KP, Novakovic B, Saffery R. Immuno-epigenomic analysis identifies attenuated interferon responses in naïve CD4 T cells of adolescents with peanut and multi-food allergy. Pediatr Allergy Immunol 2022; 33:e13890. [PMID: 36433861 DOI: 10.1111/pai.13890] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 10/26/2022] [Accepted: 11/02/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND IgE-mediated food allergies have been linked to suboptimal naïve CD4 T (nCD4T) cell activation in infancy, underlined by epigenetic and transcriptomic variation. Similar attenuated nCD4T cell activation in adolescents with food allergy have also been reported, but these are yet to be linked to specific epigenetic or transcriptional changes. METHODS We generated genome-wide DNA methylation data in purified nCD4 T cells at quiescence and following activation in a cohort of adolescents (aged 10-15 years old) with peanut allergy (peanut only or peanut + ≥1 additional food allergy) (FA, n = 29), and age-matched non-food allergic controls (NA, n = 18). Additionally, we assessed transcriptome-wide gene expression and cytokine production in these cells following activation. RESULTS We found widespread changes in DNA methylation in both NA and FA nCD4T cells in response to activation, associated with the T cell receptor signaling pathway. Adolescents with FA exhibit unique DNA methylation signatures at quiescence and post-activation at key genes involved in Th1/Th2 differentiation (RUNX3, RXRA, NFKB1A, IL4R), including a differentially methylated region (DMR) at the TNFRSF6B promoter, linked to Th1 proliferation. Combined analysis of DNA methylation, transcriptomic data and cytokine output in the same samples identified an attenuated interferon response in nCD4T cells from FA individuals following activation, with decreased expression of several interferon genes, including IFN-γ and a DMR at a key downstream gene, BST2. CONCLUSION We find that attenuated nCD4T cell responses from adolescents with food allergy are associated with specific epigenetic variation, including disruption of interferon responses, indicating dysregulation of key immune pathways that may contribute to a persistent FA phenotype. However, we recognize the small sample size, and the consequent restraint on reporting adjusted p-value statistics as limitations of the study. Further study is required to validate these findings.
Collapse
Affiliation(s)
- Samira Imran
- Murdoch Children's Research Institute, and Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Melanie R Neeland
- Murdoch Children's Research Institute, and Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Stephen Peng
- Murdoch Children's Research Institute, and Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Amanda Vlahos
- Murdoch Children's Research Institute, and Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Parkville, Victoria, Australia
| | - David Martino
- Murdoch Children's Research Institute, and Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Parkville, Victoria, Australia.,Telethon Kids Institute, University of Western, Perth, Nedlands, Australia
| | - Shyamali C Dharmage
- Murdoch Children's Research Institute, and Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Parkville, Victoria, Australia.,Allergy and Lung Health Unit, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Mimi L K Tang
- Murdoch Children's Research Institute, and Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Parkville, Victoria, Australia.,Department of Allergy and Immunology, Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Susan Sawyer
- Murdoch Children's Research Institute, and Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Parkville, Victoria, Australia.,Centre for Adolescent Health, Royal Children's Hospital Melbourne, Melbourne, Victoria, Australia
| | - Thanh D Dang
- Murdoch Children's Research Institute, and Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Vicki McWilliam
- Murdoch Children's Research Institute, and Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Parkville, Victoria, Australia.,Department of Allergy and Immunology, Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Rachel L Peters
- Murdoch Children's Research Institute, and Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Jennifer J Koplin
- Murdoch Children's Research Institute, and Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Kirsten P Perrett
- Murdoch Children's Research Institute, and Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Parkville, Victoria, Australia.,Department of Allergy and Immunology, Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Boris Novakovic
- Murdoch Children's Research Institute, and Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Richard Saffery
- Murdoch Children's Research Institute, and Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Parkville, Victoria, Australia
| |
Collapse
|
7
|
Ashley SE, Jones AC, Anderson D, Holt PG, Bosco A, Tang MLK. Remission of peanut allergy is associated with rewiring of allergen-driven T helper 2-related gene networks. Allergy 2022; 77:3015-3027. [PMID: 35615783 PMCID: PMC9790273 DOI: 10.1111/all.15324] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 02/27/2022] [Accepted: 03/15/2022] [Indexed: 01/28/2023]
Abstract
BACKGROUND The immunological changes underpinning acquisition of remission (also called sustained unresponsiveness) following food immunotherapy remain poorly defined. Limited access to effective therapies and biosamples from treatment responders has prevented progress. Probiotic peanut oral immunotherapy is highly effective at inducing remission, providing an opportunity to investigate immune changes. METHODS Using a systems biology approach, we examined gene co-expression network patterns in peanut-specific CD4+ T cell responses before and after probiotic and peanut oral immunotherapy in subjects enrolled in the PPOIT-001 randomized trial: Responders who attained remission (n = 16), placebo-treated who remained allergic (n = 16). RESULTS Acquisition of remission was associated with rewiring of gene network patterns, which was characterized by integration of T helper 2 and interferon signalling modules, markedly reduced T helper 2 gene connectivity, and shutdown in co-expression activity between T helper 2 effectors and cell cycle regulators. CONCLUSION The immunological changes underlying remission following peanut oral immunotherapy are mediated by reprogramming of T helper 2-associated gene networks in the CD4+ T cell compartment. Findings provide insight into immune mechanisms driving the acquisition of remission following oral immunotherapy, paving the way for the development of improved approaches to induce remission/sustained unresponsiveness in patients with food allergy.
Collapse
Affiliation(s)
- Sarah E. Ashley
- Allergy ImmunologyMurdoch Children’s Research InstituteMelbourneVictoriaAustralia,Department of PaediatricsThe University of MelbourneMelbourneVictoriaAustralia,The Royal Children’s HospitalMelbourneVictoriaAustralia
| | - Anya C. Jones
- Telethon Kids InstitutePerthWestern AustraliaAustralia,The University of Western AustraliaPerthWestern AustraliaAustralia
| | - Denise Anderson
- Telethon Kids InstitutePerthWestern AustraliaAustralia,The University of Western AustraliaPerthWestern AustraliaAustralia
| | - Patrick G. Holt
- Telethon Kids InstitutePerthWestern AustraliaAustralia,The University of Western AustraliaPerthWestern AustraliaAustralia
| | - Anthony Bosco
- Telethon Kids InstitutePerthWestern AustraliaAustralia,The University of Western AustraliaPerthWestern AustraliaAustralia
| | - Mimi L. K. Tang
- Allergy ImmunologyMurdoch Children’s Research InstituteMelbourneVictoriaAustralia,Department of PaediatricsThe University of MelbourneMelbourneVictoriaAustralia,The Royal Children’s HospitalMelbourneVictoriaAustralia
| |
Collapse
|
8
|
Study Protocol for a Randomised Controlled Trial Investigating the Effects of Maternal Prebiotic Fibre Dietary Supplementation from Mid-Pregnancy to Six Months’ Post-Partum on Child Allergic Disease Outcomes. Nutrients 2022; 14:nu14132753. [PMID: 35807933 PMCID: PMC9268759 DOI: 10.3390/nu14132753] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 02/01/2023] Open
Abstract
Infant allergy is the most common early manifestation of an increasing propensity for inflammation and immune dysregulation in modern environments. Refined low-fibre diets are a major risk for inflammatory diseases through adverse effects on the composition and function of gut microbiota. This has focused attention on the potential of prebiotic dietary fibres to favourably change gut microbiota, for local and systemic anti-inflammatory effects. In pregnancy, the immunomodulatory effects of prebiotics may also have benefits for the developing fetal immune system, and provide a potential dietary strategy to reduce the risk of allergic disease. Here, we present the study protocol for a double-blinded, randomised controlled trial investigating the effects of maternal prebiotics supplementation on child allergic disease outcomes. Eligible pregnant women have infants with a first-degree relative with a history of medically diagnosed allergic disease. Consented women are randomised to consume either prebiotics (galacto-oligosaccharides and fructo-oligosaccharides) or placebo (maltodextrin) powder daily from 18–20 weeks’ gestation to six months’ post-partum. The target sample size is 652 women. The primary outcome is infant medically diagnosed eczema; secondary outcomes include allergen sensitisation, food allergies and recurrent wheeze. Breast milk, stool and blood samples are collected at multiple timepoints for further analysis.
Collapse
|
9
|
Eapen AA, Parameswaran S, Forney C, Edsall LE, Miller D, Donmez O, Dunn K, Lu X, Granitto M, Rowden H, Magier AZ, Pujato M, Chen X, Kaufman K, Bernstein DI, Devonshire AL, Rothenberg ME, Weirauch MT, Kottyan LC. Epigenetic and transcriptional dysregulation in CD4+ T cells in patients with atopic dermatitis. PLoS Genet 2022; 18:e1009973. [PMID: 35576187 PMCID: PMC9135339 DOI: 10.1371/journal.pgen.1009973] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 05/26/2022] [Accepted: 04/20/2022] [Indexed: 12/30/2022] Open
Abstract
Atopic dermatitis (AD) is one of the most common skin disorders among children. Disease etiology involves genetic and environmental factors, with 29 independent AD risk loci enriched for risk allele-dependent gene expression in the skin and CD4+ T cell compartments. We investigated the potential epigenetic mechanisms responsible for the genetic susceptibility of CD4+ T cells. To understand the differences in gene regulatory activity in peripheral blood T cells in AD, we measured chromatin accessibility (an assay based on transposase-accessible chromatin sequencing, ATAC-seq), nuclear factor kappa B subunit 1 (NFKB1) binding (chromatin immunoprecipitation with sequencing, ChIP-seq), and gene expression levels (RNA-seq) in stimulated CD4+ T cells from subjects with active moderate-to-severe AD, as well as in age-matched non-allergic controls. Open chromatin regions in stimulated CD4+ T cells were highly enriched for AD genetic risk variants, with almost half of the AD risk loci overlapping AD-dependent ATAC-seq peaks. AD-specific open chromatin regions were strongly enriched for NF-κB DNA-binding motifs. ChIP-seq identified hundreds of NFKB1-occupied genomic loci that were AD- or control-specific. As expected, the AD-specific ChIP-seq peaks were strongly enriched for NF-κB DNA-binding motifs. Surprisingly, control-specific NFKB1 ChIP-seq peaks were not enriched for NFKB1 motifs, but instead contained motifs for other classes of human transcription factors, suggesting a mechanism involving altered indirect NFKB1 binding. Using DNA sequencing data, we identified 63 instances of altered genotype-dependent chromatin accessibility at 36 AD risk variant loci (30% of AD risk loci) that might lead to genotype-dependent gene expression. Based on these findings, we propose that CD4+ T cells respond to stimulation in an AD-specific manner, resulting in disease- and genotype-dependent chromatin accessibility alterations involving NFKB1 binding.
Collapse
Affiliation(s)
- Amy A. Eapen
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Division of Allergy and Clinical Immunology, Henry Ford Health System, Detroit, Michigan, United States of America
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Sreeja Parameswaran
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Carmy Forney
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Lee E. Edsall
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Daniel Miller
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Omer Donmez
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Katelyn Dunn
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Xiaoming Lu
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Marissa Granitto
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Hope Rowden
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Adam Z. Magier
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Mario Pujato
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Xiaoting Chen
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Kenneth Kaufman
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Divisions of Biomedical Informatics and Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
- Cincinnati Veterans Administration, Cincinnati, Ohio, United States of America
| | - David I. Bernstein
- Division of Immunology, Allergy, and Rheumatology, University of Cincinnati, College of Medicine, Cincinnati, Ohio, United States of America
| | - Ashley L. Devonshire
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Marc E. Rothenberg
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Matthew T. Weirauch
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Divisions of Biomedical Informatics and Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Leah C. Kottyan
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| |
Collapse
|
10
|
Lee KH, Bosco A, O'Sullivan M, Song Y, Metcalfe J, Yu K, Mullins BJ, Loh R, Zhang G. Identifying gene network patterns and associated cellular immune responses in children with or without nut allergy. World Allergy Organ J 2022; 15:100631. [PMID: 35228856 PMCID: PMC8844301 DOI: 10.1016/j.waojou.2022.100631] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 11/23/2021] [Accepted: 01/18/2022] [Indexed: 11/17/2022] Open
Abstract
Background Although evidence suggests that the immune system plays a key role in the pathophysiology of nut allergy, the precise immunological mechanisms of nut allergy have not been systematically investigated. The aim of the present study was to identify gene network patterns and associated cellular immune responses in children with or without nut allergy. Methods Transcriptome profiling of whole blood cells was compared between children with and without nut allergy. Three genes were selected to be validated on a larger cohort of samples (n = 86) by reverse transcription-polymerase chain reactions (RT-qPCR). The composition of immune cells was inferred from the transcriptomic data using the CIBERSORTx algorithm. A co-expression network was constructed employing weighted gene co-expression network analysis (WGCNA) on the top 5000 most variable transcripts. The modules were interrogated with pathway analysis tools (InnateDB) and correlated with clinical phenotypes and cellular immune responses. Results Proportions of neutrophils were positively correlated and CD4+ T-cells and regulatory T-cells (Tregs) were negatively correlated with modules of nut allergy. We also identified 2 upregulated genes, namely Interferon Induced With Helicase C Domain 1 (IFIH1), DNA damage-regulated autophagy modulator 1 (DRAM1) and a downregulated gene Zinc Finger Protein 512B (ZNF512B) as hub genes for nut allergy. Further pathway analysis showed enrichment of type 1 interferon signalling in nut allergy. Conclusions Our findings suggest that upregulation of type 1 interferon signalling and neutrophil responses and downregulation of CD4+ T-cells and Tregs are features of the pathogenesis of nut allergy.
Collapse
Affiliation(s)
- Khui Hung Lee
- School of Public Health, Curtin University of Technology, Bentley, 6102, Western Australia, Australia
| | - Anthony Bosco
- Telethon Kids Institute, University of Western Australia, Crawley, 6000, Western Australia, Australia
| | - Michael O'Sullivan
- Department of Immunology, Perth Children's Hospital, Nedlands, 6009, Western Australia, Australia
| | - Yong Song
- The Menzies Institute for Medical Research, University of Tasmania, Hobart, 7000, Tasmania, Australia
| | - Jessica Metcalfe
- Department of Immunology, Perth Children's Hospital, Nedlands, 6009, Western Australia, Australia
| | - Kan Yu
- School of Science, Edith Cowan University, Joondalup, 6027, Western Australia, Australia
| | - Benjamin J. Mullins
- School of Public Health, Curtin University of Technology, Bentley, 6102, Western Australia, Australia
| | - Richard Loh
- Department of Immunology, Perth Children's Hospital, Nedlands, 6009, Western Australia, Australia
| | - Guicheng Zhang
- School of Public Health, Curtin University of Technology, Bentley, 6102, Western Australia, Australia
- Curtin Health Innovation Research Institute, Curtin University, Bentley, 6102, Western Australia, Australia
- Infection and Immunity, School of Biomedical Sciences, University of Western Australia, Crawley, 6000, Western Australia, Australia
- Corresponding author. School of Public Health, Curtin University of Technology, Kent St, Bentley, 6102, Western Australia, Australia.
| |
Collapse
|
11
|
Devonshire AL, Fan H, Pujato M, Paranjpe A, Gursel D, Schipma M, Dunn JM, Andorf S, Pongracic JA, Kottyan LC, Kumar R. Whole blood transcriptomics identifies gene expression associated with peanut allergy in infants at high risk. Clin Exp Allergy 2021; 51:1396-1400. [PMID: 34473385 PMCID: PMC9290487 DOI: 10.1111/cea.14008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/08/2021] [Indexed: 12/16/2022]
Affiliation(s)
- Ashley L Devonshire
- Division of Allergy and Immunology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA.,Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Hanli Fan
- Pathology Core Facility, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Mario Pujato
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Aditi Paranjpe
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Demirkan Gursel
- Pathology Core Facility, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Matthew Schipma
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Julia M Dunn
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Sandra Andorf
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jacqueline A Pongracic
- Division of Allergy and Immunology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA.,Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Leah C Kottyan
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Rajesh Kumar
- Division of Allergy and Immunology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA.,Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
12
|
Imran S, Neeland MR, Koplin J, Dharmage S, Tang MLK, Sawyer S, Dang T, McWilliam V, Peters R, Perrett KP, Novakovic B, Saffery R. Epigenetic programming underpins B-cell dysfunction in peanut and multi-food allergy. Clin Transl Immunology 2021; 10:e1324. [PMID: 34466226 PMCID: PMC8384135 DOI: 10.1002/cti2.1324] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/07/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Rates of IgE-mediated food allergy (FA) have increased over the last few decades, and mounting evidence implicates disruption of epigenetic profiles in various immune cell types in FA development. Recent data implicate B-cell dysfunction in FA; however, few studies have examined epigenetic changes within these cells. METHODS We assessed epigenetic and transcriptomic profiles in purified B cells from adolescents with FA, comparing single-food-allergic (peanut only), multi-food-allergic (peanut and ≥1 other food) and non-allergic (control) individuals. Adolescents represent a phenotype of persistent and severe FA indicative of a common immune deviation. RESULTS We identified 144 differentially methylated probes (DMPs) and 116 differentially expressed genes (DEGs) that distinguish B cells of individuals with FA from controls, including differential methylation of the PM20D1 promoter previously associated with allergic disorders. Subgroup comparisons found 729 DMPs specific to either single-food- or multi-food-allergic individuals, suggesting epigenetic distinctions between allergy groups. This included two regions with increased methylation near three S100 genes in multi-food-allergic individuals. Ontology results of DEGs specific to multi-food-allergic individuals revealed enrichment of terms associated with myeloid cell activation. Motif enrichment analysis of promoters associated with DMPs and DEGs showed differential enrichment for motifs recognised by transcription factors regulating B- and T-cell development, B-cell lineage determination and TGF-β signalling pathway between the multi-food-allergic and single-food-allergic groups. CONCLUSION Our data highlight epigenetic changes in B cells associated with peanut allergy, distinguishing features of the epigenome between single-food- and multi-food-allergic individuals and revealing differential developmental pathways potentially underpinning these distinct phenotypes.
Collapse
Affiliation(s)
- Samira Imran
- Murdoch Children’s Research Institute, and Department of PaediatricsUniversity of MelbourneRoyal Children's HospitalParkvilleVICAustralia
| | - Melanie R Neeland
- Murdoch Children’s Research Institute, and Department of PaediatricsUniversity of MelbourneRoyal Children's HospitalParkvilleVICAustralia
| | - Jennifer Koplin
- Murdoch Children’s Research Institute, and Department of PaediatricsUniversity of MelbourneRoyal Children's HospitalParkvilleVICAustralia
| | - Shyamali Dharmage
- Murdoch Children’s Research Institute, and Department of PaediatricsUniversity of MelbourneRoyal Children's HospitalParkvilleVICAustralia
- Allergy and Lung Health UnitMelbourne School of Population and Global HealthUniversity of MelbourneCarltonVICAustralia
| | - Mimi LK Tang
- Murdoch Children’s Research Institute, and Department of PaediatricsUniversity of MelbourneRoyal Children's HospitalParkvilleVICAustralia
- Department of Allergy and ImmunologyRoyal Children's HospitalMelbourneVICAustralia
| | - Susan Sawyer
- Murdoch Children’s Research Institute, and Department of PaediatricsUniversity of MelbourneRoyal Children's HospitalParkvilleVICAustralia
- Centre for Adolescent HealthRoyal Children's HospitalMelbourneVICAustralia
| | - Thanh Dang
- Murdoch Children’s Research Institute, and Department of PaediatricsUniversity of MelbourneRoyal Children's HospitalParkvilleVICAustralia
| | - Vicki McWilliam
- Murdoch Children’s Research Institute, and Department of PaediatricsUniversity of MelbourneRoyal Children's HospitalParkvilleVICAustralia
- Department of Allergy and ImmunologyRoyal Children's HospitalMelbourneVICAustralia
| | - Rachel Peters
- Murdoch Children’s Research Institute, and Department of PaediatricsUniversity of MelbourneRoyal Children's HospitalParkvilleVICAustralia
| | - Kirsten P Perrett
- Murdoch Children’s Research Institute, and Department of PaediatricsUniversity of MelbourneRoyal Children's HospitalParkvilleVICAustralia
- Department of Allergy and ImmunologyRoyal Children's HospitalMelbourneVICAustralia
| | - Boris Novakovic
- Murdoch Children’s Research Institute, and Department of PaediatricsUniversity of MelbourneRoyal Children's HospitalParkvilleVICAustralia
| | - Richard Saffery
- Murdoch Children’s Research Institute, and Department of PaediatricsUniversity of MelbourneRoyal Children's HospitalParkvilleVICAustralia
| |
Collapse
|
13
|
Mapping the landscape of chromatin dynamics during naïve CD4+ T-cell activation. Sci Rep 2021; 11:14101. [PMID: 34238961 PMCID: PMC8266878 DOI: 10.1038/s41598-021-93509-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 06/22/2021] [Indexed: 12/23/2022] Open
Abstract
T-cell activation induces context-specific gene expression programs that promote energy generation and biosynthesis, progression through the cell cycle and ultimately cell differentiation. The aim of this study was to apply the omni ATAC-seq method to characterize the landscape of chromatin changes induced by T-cell activation in mature naïve CD4+ T-cells. Using a well-established ex vivo protocol of canonical T-cell receptor signaling, we generated genome-wide chromatin maps of naïve T-cells from pediatric donors in quiescent or recently activated states. We identified thousands of individual chromatin accessibility peaks that are associated with T-cell activation, the majority of which were annotated intronic and intergenic enhancer regions. A core set of 3268 gene promoters underwent chromatin remodeling and concomitant changes in gene expression in response to activation, and were enriched in multiple pathways controlling cell cycle regulation, metabolism, inflammatory response genes and cell survival. Leukemia inhibitory factor (LIF) was among those factors that gained the highest accessibility and expression, in addition to IL2-STAT5 dependent chromatin remodeling in the T-cell activation response. Using publicly available data we found the chromatin response was far more dynamic at 24-h compared with 72-h post-activation. In total 546 associations were reproduced at both time-points with similar strength of evidence and directionality of effect. At the pathways level, the IL2-STAT5, KRAS signalling and UV response pathways were replicable at both time-points, although differentially modulated from 24 to 72 h post-activation.
Collapse
|
14
|
Iweala OI, Choudhary SK, Addison CT, Commins SP. T and B Lymphocyte Transcriptional States Differentiate between Sensitized and Unsensitized Individuals in Alpha-Gal Syndrome. Int J Mol Sci 2021; 22:ijms22063185. [PMID: 33804792 PMCID: PMC8003943 DOI: 10.3390/ijms22063185] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/08/2021] [Accepted: 03/17/2021] [Indexed: 12/12/2022] Open
Abstract
The mechanisms of pathogenesis driving alpha-gal syndrome (AGS) are not fully understood. Differences in immune gene expression between AGS individuals and non-allergic controls may illuminate molecular pathways and targets critical for AGS development. We performed immune expression profiling with RNA from the peripheral blood mononuclear cells (PBMCs) of seven controls, 15 AGS participants, and two participants sensitized but not allergic to alpha-gal using the NanoString nCounter PanCancer immune profiling panel, which includes 770 genes from 14 different cell types. The top differentially expressed genes (DEG) between AGS subjects and controls included transcription factors regulating immune gene expression, such as the NFκB pathway (NFKBIA, NFKB2, REL), antigen presentation molecules, type 2/allergic immune responses, itch, and allergic dermatitis. The differential expression of genes linked to T and B cell function was also identified, including transcription factor BCL-6, markers of antigen experience (CD44) and memory (CD27), chemokine receptors (CXCR3, CXCR6), and regulators of B-cell proliferation, cell cycle entry and immunoglobulin production (CD70). The PBMCs from AGS subjects also had increased TNF and IFN-gamma mRNA expression compared to controls. AGS is associated with a distinct gene expression profile in circulating PBMCs. DEGs related to antigen presentation, antigen-experienced T-cells, and type 2 immune responses may promote the development of alpha-gal specific IgE and the maintenance of AGS.
Collapse
Affiliation(s)
- Onyinye I. Iweala
- Department of Pediatrics, University of North Carolina Food Allergy Initiative, Division of Allergy, Immunology and Rheumatology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.K.C.); (C.T.A.); (S.P.C.)
- Thurston Arthritis Research Center, Division of Rheumatology, Allergy, and Immunology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Correspondence:
| | - Shailesh K. Choudhary
- Department of Pediatrics, University of North Carolina Food Allergy Initiative, Division of Allergy, Immunology and Rheumatology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.K.C.); (C.T.A.); (S.P.C.)
- Thurston Arthritis Research Center, Division of Rheumatology, Allergy, and Immunology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Claire T. Addison
- Department of Pediatrics, University of North Carolina Food Allergy Initiative, Division of Allergy, Immunology and Rheumatology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.K.C.); (C.T.A.); (S.P.C.)
- Thurston Arthritis Research Center, Division of Rheumatology, Allergy, and Immunology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Scott P. Commins
- Department of Pediatrics, University of North Carolina Food Allergy Initiative, Division of Allergy, Immunology and Rheumatology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.K.C.); (C.T.A.); (S.P.C.)
- Thurston Arthritis Research Center, Division of Rheumatology, Allergy, and Immunology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
15
|
Neeland MR, Andorf S, Dang TD, McWilliam VL, Perrett KP, Koplin JJ, Saffery R. Altered immune cell profiles and impaired CD4 T-cell activation in single and multi-food allergic adolescents. Clin Exp Allergy 2021; 51:674-684. [PMID: 33626189 DOI: 10.1111/cea.13857] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/10/2021] [Accepted: 02/17/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Approximately 5% of adolescents have a food allergy, with peanut and tree nut allergies the most common. Having two or more food allergies in adolescence also doubles the risk of any adverse food reaction, and is associated with increased dietary and social burden. Investigations of immune function in persistently food allergic children are rare. OBJECTIVE In the present study, we aimed to investigate the immune mechanisms that underlie food allergy in adolescence. METHODS We used high-dimensional flow cytometry, unsupervised computational analysis and functional studies to comprehensively phenotype a range of non-antigen-specific immune parameters in a group of well-characterized adolescents with clinically defined single peanut allergy, multi-food allergy and aged-matched non-food allergic controls. RESULTS We show that food allergic adolescents have higher circulating proportions of dendritic cells (p = .0084, FDR-adjusted p = .087, median in no FA: 0.63% live cells, in FA: 0.93%), and higher frequency of activated, memory-like Tregs relative to non-food allergic adolescents (p = .011, FDR-adjusted p = .087, median in no FA: 0.49% live cells, in FA: 0.65%). Cytokine profiling revealed that CD3/CD28 stimulated naïve CD4 T cells from food allergic adolescents produced less IL-6 (p = .0020, FDR-adjusted p = .018, median log2 fold change [stimulated/unstimulated] in no FA: 3.03, in FA: 1.92) and TNFα (p = .0044, FDR-adjusted p = .020, median in no FA: 9.16, in FA: 8.64) and may secrete less IFNγ (p = .035, FDR-adjusted p = .11, median in no FA: 6.29, in FA: 5.67) than naïve CD4 T cells from non-food allergic controls. No differences between clinical groups were observed for LPS-stimulated monocyte secretion of cytokines. CONCLUSIONS These results have important implications for understanding the evolution of the immune response in food allergy throughout childhood, revealing that dendritic cell and T-cell signatures previously identified in early life may persist through to adolescence.
Collapse
Affiliation(s)
- Melanie R Neeland
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Vic, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, Vic, Australia
| | - Sandra Andorf
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Divisions of Biomedical Informatics and Allergy & Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Thanh D Dang
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Vic, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, Vic, Australia
| | - Vicki L McWilliam
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Vic, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, Vic, Australia
| | - Kirsten P Perrett
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Vic, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, Vic, Australia.,Department of Allergy and Immunology, Royal Children's Hospital, Melbourne, Vic, Australia
| | - Jennifer J Koplin
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Vic, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, Vic, Australia
| | - Richard Saffery
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Vic, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, Vic, Australia
| |
Collapse
|
16
|
Patil SU, Bunyavanich S, Cecilia Berin M. Emerging Food Allergy Biomarkers. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2020; 8:2516-2524. [PMID: 32888527 PMCID: PMC7479640 DOI: 10.1016/j.jaip.2020.04.054] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/17/2020] [Accepted: 04/18/2020] [Indexed: 12/12/2022]
Abstract
The management of food allergy is complicated by the lack of highly predictive biomarkers for diagnosis and prediction of disease course. The measurement of food-specific IgE is a useful tool together with clinical history but is an imprecise predictor of clinical reactivity. The gold standard for diagnosis and clinical research is a double-blind placebo-controlled food challenge. Improvement in our understanding of immune mechanisms of disease, development of high-throughput technologies, and advances in bioinformatics have yielded a number of promising new biomarkers of food allergy. In this review, we will discuss advances in immunoglobulin measurements, the utility of the basophil activation test, T-cell profiling, and the use of -omic technologies (transcriptome, epigenome, microbiome, and metabolome) as biomarker tools in food allergy.
Collapse
Affiliation(s)
- Sarita U. Patil
- Food Allergy Center, Department of Pediatrics, Massachusetts General Hospital, Boston, MA 02114
- Center for Immunological and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Supinda Bunyavanich
- Jaffe Food Allergy Institute, Department of Pediatrics; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - M. Cecilia Berin
- Jaffe Food Allergy Institute, Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| |
Collapse
|
17
|
Do AN, Watson CT, Cohain AT, Griffin RS, Grishin A, Wood RA, Wesley Burks A, Jones SM, Scurlock A, Leung DYM, Sampson HA, Sicherer SH, Sharp AJ, Schadt EE, Bunyavanich S. Dual transcriptomic and epigenomic study of reaction severity in peanut-allergic children. J Allergy Clin Immunol 2020; 145:1219-1230. [PMID: 31838046 PMCID: PMC7192362 DOI: 10.1016/j.jaci.2019.10.040] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 09/27/2019] [Accepted: 10/29/2019] [Indexed: 12/15/2022]
Abstract
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.
Collapse
Affiliation(s)
- Anh N Do
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Corey T Watson
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, Ky
| | - Ariella T Cohain
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Robert S Griffin
- Department of Anesthesia, Hospital for Special Surgery, New York, NY
| | - Alexander Grishin
- Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Robert A Wood
- Department of Pediatrics, Johns Hopkins University, Baltimore, Md
| | - A Wesley Burks
- Department of Pediatrics, University of North Carolina, Chapel Hill, NC
| | - Stacie M Jones
- Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock, Ark
| | - Amy Scurlock
- Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock, Ark
| | | | - Hugh A Sampson
- Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Scott H Sicherer
- Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Andrew J Sharp
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Eric E Schadt
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Supinda Bunyavanich
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY; Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
18
|
Saglani S, Gregory LG, Manghera AK, Branchett WJ, Uwadiae F, Entwistle LJ, Oliver RA, Vasiliou JE, Sherburn R, Lui S, Puttur F, Vöhringer D, Walker SA, Buckley J, Grychtol R, Fainardi V, Denney L, Byrne A, von Mutius E, Bush A, Lloyd CM. Inception of early-life allergen-induced airway hyperresponsiveness is reliant on IL-13 +CD4 + T cells. Sci Immunol 2019; 3:3/27/eaan4128. [PMID: 30194239 DOI: 10.1126/sciimmunol.aan4128] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 07/09/2018] [Indexed: 12/15/2022]
Abstract
Airway hyperresponsiveness (AHR) is a critical feature of wheezing and asthma in children, but the initiating immune mechanisms remain unconfirmed. We demonstrate that both recombinant interleukin-33 (rIL-33) and allergen [house dust mite (HDM) or Alternaria alternata] exposure from day 3 of life resulted in significantly increased pulmonary IL-13+CD4+ T cells, which were indispensable for the development of AHR. In contrast, adult mice had a predominance of pulmonary LinnegCD45+CD90+IL-13+ type 2 innate lymphoid cells (ILC2s) after administration of rIL-33. HDM exposure of neonatal IL-33 knockout (KO) mice still resulted in AHR. However, neonatal CD4creIL-13 KO mice (lacking IL-13+CD4+ T cells) exposed to allergen from day 3 of life were protected from AHR despite persistent pulmonary eosinophilia, elevated IL-33 levels, and IL-13+ ILCs. Moreover, neonatal mice were protected from AHR when inhaled Acinetobacter lwoffii (an environmental bacterial isolate found in cattle farms, which is known to protect from childhood asthma) was administered concurrent with HDM. A. lwoffii blocked the expansion of pulmonary IL-13+CD4+ T cells, whereas IL-13+ ILCs and IL-33 remained elevated. Administration of A. lwoffii mirrored the findings from the CD4creIL-13 KO mice, providing a translational approach for disease protection in early life. These data demonstrate that IL-13+CD4+ T cells, rather than IL-13+ ILCs or IL-33, are critical for inception of allergic AHR in early life.
Collapse
Affiliation(s)
- Sejal Saglani
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK. .,Respiratory Paediatrics, Royal Brompton Hospital and National Heart and Lung Institute, Imperial College London, London, UK
| | - Lisa G Gregory
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - Avneet K Manghera
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - William J Branchett
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - Faith Uwadiae
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - Lewis J Entwistle
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - R A Oliver
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - Jessica E Vasiliou
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - Rebekah Sherburn
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - Stephen Lui
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - F Puttur
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - David Vöhringer
- Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg Wasserturmstrasse 3-5, 91054 Erlangen, Germany
| | - Simone A Walker
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - James Buckley
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - Ruth Grychtol
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - Valentina Fainardi
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - Laura Denney
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - Adam Byrne
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - Erika von Mutius
- Dr Von Hauner Childrens Hospital of Ludwig-Maximilians-Universitaet of Munich and Helmholtz Zentrum Munchen-German Research Center for Environmental Health, Institute for Asthma and Allergy Prevention, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
| | - Andrew Bush
- Respiratory Paediatrics, Royal Brompton Hospital and National Heart and Lung Institute, Imperial College London, London, UK
| | - Clare M Lloyd
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
19
|
Epigenetic dysregulation of naive CD4+ T-cell activation genes in childhood food allergy. Nat Commun 2018; 9:3308. [PMID: 30120223 PMCID: PMC6098117 DOI: 10.1038/s41467-018-05608-4] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 06/05/2018] [Indexed: 12/31/2022] Open
Abstract
Food allergy poses a significant clinical and public health burden affecting 2–10% of infants. Using integrated DNA methylation and transcriptomic profiling, we found that polyclonal activation of naive CD4+ T cells through the T cell receptor results in poorer lymphoproliferative responses in children with immunoglobulin E (IgE)-mediated food allergy. Reduced expression of cell cycle-related targets of the E2F and MYC transcription factor networks, and remodeling of DNA methylation at metabolic (RPTOR, PIK3D, MAPK1, FOXO1) and inflammatory genes (IL1R, IL18RAP, CD82) underpins this suboptimal response. Infants who fail to resolve food allergy in later childhood exhibit cumulative increases in epigenetic disruption at T cell activation genes and poorer lymphoproliferative responses compared to children who resolved food allergy. Our data indicate epigenetic dysregulation in the early stages of signal transduction through the T cell receptor complex, and likely reflects pathways modified by gene–environment interactions in food allergy. Immunoglobulin E (IgE)-mediated food allergy is a major issue that affects 2–10% of infants. Here the authors study the epigenetic regulation of the naive CD4+ T cell activation response among children with IgE-mediated food allergy finding epigenetic dysregulation in the early stages of signal transduction through the T cell receptor complex.
Collapse
|
20
|
Watson CT, Cohain AT, Griffin RS, Chun Y, Grishin A, Hacyznska H, Hoffman GE, Beckmann ND, Shah H, Dawson P, Henning A, Wood R, Burks AW, Jones SM, Leung DYM, Sicherer S, Sampson HA, Sharp AJ, Schadt EE, Bunyavanich S. Integrative transcriptomic analysis reveals key drivers of acute peanut allergic reactions. Nat Commun 2017; 8:1943. [PMID: 29203772 PMCID: PMC5715016 DOI: 10.1038/s41467-017-02188-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 11/09/2017] [Indexed: 02/07/2023] Open
Abstract
Mechanisms driving acute food allergic reactions have not been fully characterized. We profile the dynamic transcriptome of acute peanut allergic reactions using serial peripheral blood samples obtained from 19 children before, during, and after randomized, double-blind, placebo-controlled oral challenges to peanut. We identify genes with changes in expression triggered by peanut, but not placebo, during acute peanut allergic reactions. Network analysis reveals that these genes comprise coexpression networks for acute-phase response and pro-inflammatory processes. Key driver analysis identifies six genes (LTB4R, PADI4, IL1R2, PPP1R3D, KLHL2, and ECHDC3) predicted to causally modulate the state of coregulated networks in response to peanut. Leukocyte deconvolution analysis identifies changes in neutrophil, naive CD4+ T cell, and macrophage populations during peanut challenge. Analyses in 21 additional peanut allergic subjects replicate major findings. These results highlight key genes, biological processes, and cell types that can be targeted for mechanistic study and therapeutic targeting of peanut allergy.
Collapse
Affiliation(s)
- C T Watson
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - A T Cohain
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - R S Griffin
- Department of Anesthesia, Hospital for Special Surgery, New York, NY, 10021, USA
| | - Y Chun
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - A Grishin
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - H Hacyznska
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - G E Hoffman
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - N D Beckmann
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - H Shah
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - P Dawson
- eEmmes Corporation, Rockville, MD, 20850, USA
| | - A Henning
- eEmmes Corporation, Rockville, MD, 20850, USA
| | - R Wood
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - A W Burks
- Department of Pediatrics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - S M Jones
- Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock, AS, 72202, USA
| | - D Y M Leung
- Department of Pediatrics, National Jewish Health, Denver, CO, 80206, USA
| | - S Sicherer
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - H A Sampson
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - A J Sharp
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - E E Schadt
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - S Bunyavanich
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
21
|
Bunning BJ, DeKruyff RH, Nadeau KC. Epigenetic Changes During Food-Specific Immunotherapy. Curr Allergy Asthma Rep 2017; 16:87. [PMID: 27943047 DOI: 10.1007/s11882-016-0665-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW The prevalence and severity of IgE-mediated food allergy has increased dramatically over the last 15 years and is becoming a global health problem. Multiple lines of evidence suggest that epigenetic modifications of the genome resulting from gene-environment interactions have a key role in the increased prevalence of atopic disease. In this review, we describe the recent evidence suggesting how epigenetic changes mediate susceptibility to food allergies, and discuss how immunotherapy (IT) may reverse these effects. We discuss the areas of the epigenome as yet unexplored in terms of food allergy and IT such as histone modification and chromatin accessibility, and new techniques that may be utilized in future studies. RECENT FINDINGS Recent findings provide strong evidence that DNA methylation of certain promoter regions such as Forkhead box protein 3 is associated with clinical reactivity, and further, can be changed during IT treatment. Reports on other epigenetic changes are limited but also show evidence of significant change based on both disease status and treatment. In comparison to epigenetic studies focusing on asthma and allergic rhinitis, food allergy remains understudied. However, within the next decade, it is likely that epigenetic modifications may be used as biomarkers to aid in diagnosis and treatment of food-allergic patients. DNA methylation at specific loci has shown associations between food challenge outcomes, successful desensitization treatment, and overall phenotype compared to healthy controls.
Collapse
Affiliation(s)
- Bryan J Bunning
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, CA, USA.,Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, CA, USA
| | - Rosemarie H DeKruyff
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, CA, USA.,Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, CA, USA
| | - Kari C Nadeau
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, CA, USA. .,Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, CA, USA. .,Sean N. Parker Center for Allergy and Asthma Research, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford University School of Medicine, 269 Campus Drive, CCSR 3215, MC 5366, Stanford, CA, 94305-5101, USA.
| |
Collapse
|
22
|
Stokholm J, Sevelsted A, Anderson UD, Bisgaard H. Preeclampsia Associates with Asthma, Allergy, and Eczema in Childhood. Am J Respir Crit Care Med 2017; 195:614-621. [PMID: 27626972 DOI: 10.1164/rccm.201604-0806oc] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
RATIONALE Preeclampsia reflects an unusual increase in systemic inflammation during pregnancy. OBJECTIVES We studied associations between preeclampsia and asthma, allergy, and eczema in Copenhagen Prospective Studies on Asthma in Childhood2000 (COPSAC2000) and in national registries. METHODS COPSAC2000 is a high-risk birth cohort of 411 Danish children. Asthma, allergy, and eczema were diagnosed prospectively, and lung function measured at age 1 month and 7 years. Sensitization was evaluated at age 6 months, 18 months, 4 years, and 6 years by skin prick tests and IgE measurements. The register-based cohort included 1.7 million children from Danish national registries in the 35-year period 1977-2012. Children born to mothers with preeclampsia were analyzed regarding risk of asthma, allergy, and eczema. MEASUREMENTS AND MAIN RESULTS In the COPSAC2000 cohort, 5.6% (n = 23) were diagnosed with preeclampsia. Preeclampsia was associated with increased risk of treatment with inhaled corticosteroids at age 7 years (adjusted odds ratio, 4.01 [95% confidence interval (CI), 1.11-14.43]; P = 0.0337), increased bronchial responsiveness to methacholine (adjusted β-coefficient log-μmol, -0.80 [95% CI, -1.55 to -0.06]; P = 0.0348), and allergic rhinitis (adjusted odds ratio, 4.83 [95% CI, 1.58-14.78]; P = 0.0057) in the 7-year-old children. Furthermore, the children had an increased risk of sensitization to both aeroallergens and food allergens, and increased amount of total IgE during childhood. In the registry-based cohort, 3.7% (n = 62,728) were born to mothers with preeclampsia. Preeclampsia was associated with increased risk of asthma, eczema, and aeroallergen and food allergy, especially pronounced after a duration of preeclampsia of 14 days or more. Maternal asthma increased the risk of preeclampsia. CONCLUSIONS Preeclampsia is a shared prenatal risk factor for asthma, eczema, and allergy in childhood pointing toward in utero immune programming of the child.
Collapse
Affiliation(s)
- Jakob Stokholm
- 1 Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark.,2 Department of Pediatrics, Naestved Hospital, Naestved, Denmark; and
| | - Astrid Sevelsted
- 1 Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Ulrik D Anderson
- 3 Department of Obstetrics and Gynecology, and.,4 Department of Clinical Sciences, Skåne University Hospital, Lund University, Lund, Sweden
| | - Hans Bisgaard
- 1 Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
23
|
The role of PKCζ in cord blood T-cell maturation towards Th1 cytokine profile and its epigenetic regulation by fish oil. Biosci Rep 2017; 37:BSR20160485. [PMID: 28159873 PMCID: PMC5482199 DOI: 10.1042/bsr20160485] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 01/25/2017] [Accepted: 02/03/2017] [Indexed: 01/21/2023] Open
Abstract
While immunodeficiency of immaturity of the neonate has been considered important as the basis for unusual susceptibility to infection, it has also been recognized that the ability to progress from an immature Th2 cytokine predominance to a Th1 profile has relevance in determining whether children will develop allergy, providing an opportunity for epigenetic regulation through environmental pressures. However, this notion remains relatively unexplored. Here, we present evidence that there are two major control points to explain the immunodeficiency in cord blood (CB) T-cells, a deficiency in interleukin (IL)-12 (IL-12) producing and IL-10 overproducing accessory cells, leading to a decreased interferon γ (IFNγ) synthesis and the other, an intrinsic defect in T-cell protein kinase C (PKC) ζ (PKCζ) expression. An important finding was that human CB T-cells rendered deficient in PKCζ, by shRNA knockdown, develop into low tumour necrosis factor α (TNFα) and IFNγ but increased IL-13 producing cells. Interestingly, we found that the increase in PKCζ levels in CB T-cells caused by prenatal supplementation with fish oil correlated with modifications of histone acetylation at the PKCζ gene (PRKCZ) promoter. The data demonstrate that PKCζ expression regulates the maturation of neonatal T-cells into specific functional phenotypes and that environmental influences may work via PKCζ to regulate these phenotypes and disease susceptibility.
Collapse
|
24
|
Berin MC, Shreffler WG. Mechanisms Underlying Induction of Tolerance to Foods. Immunol Allergy Clin North Am 2016; 36:87-102. [DOI: 10.1016/j.iac.2015.08.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
25
|
Fluorescence-based bioassays for the detection and evaluation of food materials. SENSORS 2015; 15:25831-67. [PMID: 26473869 PMCID: PMC4634490 DOI: 10.3390/s151025831] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 09/28/2015] [Accepted: 09/30/2015] [Indexed: 12/12/2022]
Abstract
We summarize here the recent progress in fluorescence-based bioassays for the detection and evaluation of food materials by focusing on fluorescent dyes used in bioassays and applications of these assays for food safety, quality and efficacy. Fluorescent dyes have been used in various bioassays, such as biosensing, cell assay, energy transfer-based assay, probing, protein/immunological assay and microarray/biochip assay. Among the arrays used in microarray/biochip assay, fluorescence-based microarrays/biochips, such as antibody/protein microarrays, bead/suspension arrays, capillary/sensor arrays, DNA microarrays/polymerase chain reaction (PCR)-based arrays, glycan/lectin arrays, immunoassay/enzyme-linked immunosorbent assay (ELISA)-based arrays, microfluidic chips and tissue arrays, have been developed and used for the assessment of allergy/poisoning/toxicity, contamination and efficacy/mechanism, and quality control/safety. DNA microarray assays have been used widely for food safety and quality as well as searches for active components. DNA microarray-based gene expression profiling may be useful for such purposes due to its advantages in the evaluation of pathway-based intracellular signaling in response to food materials.
Collapse
|
26
|
Abstract
It has been recognized for centuries that allergic disease runs in families, implying a role for genetic factors in determining individual susceptibility. More recently, a range of evidence shows that many of these genetic factors, together with in utero environmental exposures, lead to the development of allergic disease through altered immune and organ development. Environmental exposures during pregnancy including diet, nutrient intake and toxin exposures can alter the epigenome and interact with inherited genetic and epigenetic risk factors to directly and indirectly influence organ development and immune programming. Understanding of these factors will be essential in identifying at-risk individuals and possible development of therapeutic interventions for the primary prevention of allergic disease. In this review, we summarize the evidence that suggests allergic disease begins in utero, together with possible mechanisms for the effect of environmental exposures during pregnancy on allergic disease risk, including epigenetics.
Collapse
Affiliation(s)
- Gabrielle A Lockett
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Johanna Huoman
- Department of Clinical and Experimental Medicine, Division of Neuro and Inflammation Sciences, Unit of Autoimmunity and Immune Regulation, Linköping University, Linköping, Sweden
| | - John W Holloway
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK.,Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,International Inflammation network (in-FLAME) of the World Universities Network
| |
Collapse
|
27
|
The role of epigenetic mediation and the future of food allergy research. Semin Cell Dev Biol 2015; 43:125-130. [PMID: 26150170 DOI: 10.1016/j.semcdb.2015.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 06/29/2015] [Accepted: 07/01/2015] [Indexed: 11/22/2022]
Abstract
IgE-mediated food allergy is a developing global health problem with prevalence rising at alarmingly fast rates. In this review, we discuss the interplay between genetics, epigenetics, and environmental exposures in the pathogenesis of food allergies. We aim to highlight the most recent evidence that suggests how epigenetic control may mediate genetic susceptibility of food allergies. We also examine how epigenetic modifications may be the key in explaining how environmental factors modulate and modify gene expression, leading to the dysregulation of immune tolerance and consequently, the development of food allergies. The emerging epigenetic paradigm in food allergies is likely to provide new mechanistic insight into food allergy risk and development as well as shape our therapeutic and preventive strategies.
Collapse
|
28
|
Tsuji M. Useful biomarkers for assessing the adverse health effects of PCBs in allergic children: pediatric molecular epidemiology. Environ Health Prev Med 2015; 20:3-11. [PMID: 25344634 PMCID: PMC4284252 DOI: 10.1007/s12199-014-0419-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 09/30/2014] [Indexed: 01/09/2023] Open
Abstract
The incidences of childhood allergies have been increasing in recent years in many parts of the world. The development of allergic disorders is attributed to a complex series of interactions between individuals' genetic backgrounds and their immune and psychoneurotic responses to environmental factors. Among the various possible environmental causes of childhood allergies, the early exposure of developing infants to air pollutants and the presence of persistent chemical pollutants such as pesticides have been suggested most frequently. Therefore, it is very important to obtain epidemiological evidence of direct associations between clearly defined adverse health effects and exposure to low levels of pollutants. However, there are no useful biomarkers for assessing such associations. Thus, we planned to establish reliable health-related biomarkers that could be used to investigate these relationships in children. The serum concentrations of several sub-types of polychlorinated biphenyl (PCB) congeners were found to be significantly correlated with interleukin (IL)-8 mRNA expression among asthmatic children. In addition, IL-22 mRNA expression was found to be particularly useful for detecting the effects of environmental pollutants, especially PCB congeners, in a sub-population of vulnerable children who exhibited positive immunoglobulin E (IgE) responses to milk or egg. Furthermore, we detected significant differences in IL-22 mRNA expression between the IgE-negative non-asthmatic subjects and the asthmatic children who exhibited positive IgE reactions toward egg or milk. In conclusion, IL-8 and IL-22 mRNA expressions could be useful biomarkers for detecting sub-populations of children who are particularly vulnerable to the adverse health effects of environmental pollutants, especially PCBs.
Collapse
Affiliation(s)
- Mayumi Tsuji
- Department of Environmental Health, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyusyu, 807-8555, Japan,
| |
Collapse
|
29
|
Martino D, Joo JE, Sexton-Oates A, Dang T, Allen K, Saffery R, Prescott S. Epigenome-wide association study reveals longitudinally stable DNA methylation differences in CD4+ T cells from children with IgE-mediated food allergy. Epigenetics 2014; 9:998-1006. [PMID: 24762976 DOI: 10.4161/epi.28945] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
UNLABELLED Food allergy is mediated by a combination of genetic and environmental risk factors, potentially mediated by epigenetic mechanisms. CD4+ T-cells are key drivers of the allergic response, and may therefore harbor epigenetic variation in association with the disease phenotype. Here we retrospectively examined genome-wide DNA methylation profiles (~450,000 CpGs) from CD4+ T-cells on a birth cohort of 12 children with IgE-mediated food allergy diagnosed at 12-months, and 12 non-allergic controls. DNA samples were available at two time points, birth and 12-months. CASE control comparisons of CD4+ methylation profiles identified 179 differentially methylated probes (DMP) at 12-months and 136 DMP at birth (FDR-adjusted P value<0.05, delta β>0.1). Approximately 30% of DMPs were coincident with previously annotated SNPs. A total of 92 [corrected] allergy-associated non-SNP DMPs were present at birth when individuals were initially disease-free, potentially implicating these loci in the causal pathway. Pathway analysis of differentially methylated genes identified several MAP kinase signaling molecules. Mass spectrometry was used to validate 15 CpG sites at 3 candidate genes. Combined analysis of differential methylation with gene expression profiles revealed gene expression differences at some but not all allergy associated differentially methylated genes. Thus, dysregulation of DNA methylation at MAPK signaling-associated genes during early CD4+ T-cell development may contribute to suboptimal T-lymphocyte responses in early childhood associated with the development of food allergy.
Collapse
Affiliation(s)
- David Martino
- Murdoch Childrens Research Institute; Royal Children's Hospital; Melbourne, VIC Australia; NHMRC Centre for Food and Allergy Research; Murdoch Childrens Research Institute; Royal Children's Hospital; Melbourne, VIC Australia; Honorary Fellow University of Melbourne; Melbourne, VIC Australia
| | - Jihoon E Joo
- Murdoch Childrens Research Institute; Royal Children's Hospital; Melbourne, VIC Australia
| | - Alexandra Sexton-Oates
- Murdoch Childrens Research Institute; Royal Children's Hospital; Melbourne, VIC Australia
| | - Thanh Dang
- Murdoch Childrens Research Institute; Royal Children's Hospital; Melbourne, VIC Australia
| | - Katrina Allen
- Murdoch Childrens Research Institute; Royal Children's Hospital; Melbourne, VIC Australia; NHMRC Centre for Food and Allergy Research; Murdoch Childrens Research Institute; Royal Children's Hospital; Melbourne, VIC Australia
| | - Richard Saffery
- Murdoch Childrens Research Institute; Royal Children's Hospital; Melbourne, VIC Australia
| | - Susan Prescott
- Childhood Allergy and Immunology Research; School of Paediatrics and Child Health; University of Western Australia; Crawley, WA Australia
| |
Collapse
|
30
|
Palmer DJ, Metcalfe J, Makrides M, Gold MS, Quinn P, West CE, Loh R, Prescott SL. Early regular egg exposure in infants with eczema: A randomized controlled trial. J Allergy Clin Immunol 2013; 132:387-92.e1. [PMID: 23810152 DOI: 10.1016/j.jaci.2013.05.002] [Citation(s) in RCA: 230] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Revised: 05/12/2013] [Accepted: 05/15/2013] [Indexed: 01/08/2023]
Abstract
BACKGROUND Observational studies suggest that early regular ingestion of allergenic foods might reduce the risk of food allergy. OBJECTIVE We sought to determine whether early regular oral egg exposure will reduce subsequent IgE-mediated egg allergy in infants with moderate-to-severe eczema. METHODS In a double-blind, randomized controlled trial infants were allocated to 1 teaspoon of pasteurized raw whole egg powder (n = 49) or rice powder (n = 37) daily from 4 to 8 months of age. Cooked egg was introduced to both groups after an observed feed at 8 months. The primary outcome was IgE-mediated egg allergy at 12 months, as defined based on the results of an observed pasteurized raw egg challenge and skin prick tests. RESULTS A high proportion (31% [15/49]) of infants randomized to receive egg had an allergic reaction to the egg powder and did not continue powder ingestion. At 4 months of age, before any known egg ingestion, 36% (24/67) of infants already had egg-specific IgE levels of greater than 0.35 kilounits of antibody (kUA)/L. At 12 months, a lower (but not significant) proportion of infants in the egg group (33%) were given a diagnosis of IgE-mediated egg allergy compared with the control group (51%; relative risk, 0.65; 95% CI, 0.38-1.11; P = .11). Egg-specific IgG4 levels were significantly (P < .001) greater in the egg group at both 8 and 12 months. CONCLUSION Induction of immune tolerance pathways and reduction in egg allergy incidence can be achieved by early regular oral egg exposure in infants with eczema. Caution needs to be taken when these high-risk infants are first exposed to egg because many have sensitization already by 4 months of age.
Collapse
Affiliation(s)
- Debra J Palmer
- School of Paediatrics and Child Health, University of Western Australia, Perth, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Novakovic B, Saffery R. The ever growing complexity of placental epigenetics – Role in adverse pregnancy outcomes and fetal programming. Placenta 2012; 33:959-70. [DOI: 10.1016/j.placenta.2012.10.003] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 10/02/2012] [Accepted: 10/06/2012] [Indexed: 02/01/2023]
|
32
|
Tsuji M, Kawamoto T, Koriyama C, Matsumura F. IL-22 mRNA expression in blood samples as a useful biomarker for assessing the adverse health effects of PCBs on allergic children. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2012; 9:4321-32. [PMID: 23330224 PMCID: PMC3546764 DOI: 10.3390/ijerph9124321] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 11/15/2012] [Accepted: 11/20/2012] [Indexed: 12/11/2022]
Abstract
To facilitate the assessment of adverse effects of very low concentrations of air pollutants on general populations, we planned to establish a reliable biomarker that is also useful in identifying vulnerable populations. For this purpose we monitored several inflammation markers in blood samples from 2 year old Japanese children (N = 30), and found that those children living close to major highways (<50 m) show higher levels of mRNA expression IL-22 in their blood samples than those living further away (+50 m). This tendency was more pronounced among subjects showing positive IgE against egg and milk. We further examined association between IL-22 mRNA expression and PCB residues and found a number of significant positive correlations between each individual PCB congener and IL-22 expression. To identify the most vulnerable population among those children we selected asthma as a typical allergy-related disease, and could show that there are significant differences in the levels of IL-22 mRNA expression between IgE negative non-asthmatic subject and asthmatic children showing positive IgE reaction toward egg or milk, again. These observations support our main conclusion that IL-22 expression is a sensitive biomarker which is useful in identifying sub-populations of children who are especially vulnerable to air pollution.
Collapse
Affiliation(s)
- Mayumi Tsuji
- Department of Environmental Toxicology, University of California Davis, One Shields Ave, Davis, CA 95616, USA;
- Department of Environmental Health, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyusyu 807-8555, Japan;
| | - Toshihiro Kawamoto
- Department of Environmental Health, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyusyu 807-8555, Japan;
| | - Chihaya Koriyama
- Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8520, Japan;
| | - Fumio Matsumura
- Department of Environmental Toxicology, University of California Davis, One Shields Ave, Davis, CA 95616, USA;
| |
Collapse
|
33
|
Oral application of bacterial lysate in infancy decreases the risk of atopic dermatitis in children with 1 atopic parent in a randomized, placebo-controlled trial. J Allergy Clin Immunol 2012; 129:1040-7. [PMID: 22464674 DOI: 10.1016/j.jaci.2012.02.005] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Revised: 01/15/2012] [Accepted: 02/01/2012] [Indexed: 01/01/2023]
Abstract
BACKGROUND Lower prevalence of atopy was found in children with continuous exposure to livestock and thus to microbial compounds. In animal models exposure to endotoxin (LPS) decreases allergic sensitization and airway inflammation. OBJECTIVE We sought to evaluate the effect of orally applied bacterial lysate in infancy on the prevalence of atopic dermatitis (AD) after the treatment phase at 7 months of age. METHODS This randomized, placebo-controlled trial included 606 newborns with at least single heredity for atopy. From week 5 until the end of month 7, infants were treated orally with bacterial lysate containing heat-killed gram-negative Escherichia coli Symbio and gram-positive Enterococcus faecalis Symbio or its placebo. Children were followed until 3 years of age. RESULTS There was no difference in the primary outcome between the active and placebo groups in the total study group. AD prevalence was significantly reduced at the end of the intervention phase (31 weeks of age) in the subgroup of infants with single heredity for atopy (relative risk, 0.52; 95% CI, 0.3-0.9). Ten percent (15/154) of infants in the active group had AD compared with 19% (27/145, P = .030) in the placebo group. This was more pronounced in the group of infants with paternal heredity for atopy (11% vs 32%, P = .004; relative risk, 0.34; 95% CI, 0.2-0.7). CONCLUSION Feeding of bacterial lysate might have prevented the development of AD, especially in children with paternal atopy, possibly indicating a preventive property only in subjects with a limited risk for atopy.
Collapse
|
34
|
Martino D, Maksimovic J, Joo JH, Prescott SL, Saffery R. Genome-scale profiling reveals a subset of genes regulated by DNA methylation that program somatic T-cell phenotypes in humans. Genes Immun 2012; 13:388-98. [PMID: 22495533 DOI: 10.1038/gene.2012.7] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The aim of this study was to investigate the dynamics and relationship between DNA methylation and gene expression during early T-cell development. Mononuclear cells were collected at birth and at 12 months from 60 infants and were either activated with anti-CD3 for 24 h or cultured in media alone, and the CD4+ T-cell subset purified. DNA and RNA were co-harvested and DNA methylation was measured in 450 000 CpG sites in parallel with expression measurements taken from 25 000 genes. In unstimulated cells, we found that a subset of 1188 differentially methylated loci were associated with a change in expression in 599 genes (adjusted P value<0.01, β-fold >0.1). These genes were enriched in reprogramming regions of the genome known to control pluripotency. In contrast, over 630 genes were induced following low-level T-cell activation, but this was not associated with any significant change in DNA methylation. We conclude that DNA methylation is dynamic during early T-cell development, and has a role in the consolidation of T-cell-specific gene expression. During the early phase of clonal expansion, DNA methylation is stable and therefore appears to be of limited importance in short-term T-cell responsiveness.
Collapse
Affiliation(s)
- D Martino
- Cancer, Disease and Developmental Epigenetics, Murdoch Children's Research Institute, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | | | | | | | | |
Collapse
|
35
|
Affiliation(s)
- I. Lehmann
- Department of Environmental Immunology; UFZ - Helmholtz Centre for Environmental Research Leipzig; Leipzig; Germany
| | - G. Herberth
- Department of Environmental Immunology; UFZ - Helmholtz Centre for Environmental Research Leipzig; Leipzig; Germany
| |
Collapse
|
36
|
AIMP1 deficiency enhances airway hyperreactivity in mice via increased TH2 immune responses. Clin Immunol 2012; 143:256-65. [PMID: 22472603 DOI: 10.1016/j.clim.2012.02.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 02/10/2012] [Accepted: 02/21/2012] [Indexed: 12/22/2022]
Abstract
Aminoacyl tRNA synthetase complex-interacting multicomplex protein 1 (AIMP1) is known as a novel cytokine carrying out a variety of biological activities, including angiogenesis and wound repair. In our previous reports AIMP1 was demonstrated to induce TH1 polarization. However, the effects of AIMP1 deficiency in TH1 or TH2 immune disorders remain unclear. In this study, we characterized phenotypes of AIMP1-deficient mice and investigated the role of AIMP1 in TH2-biased airway hyperreactivity. Clinical signs of allergic airway inflammation were assessed in AIMP1-deficient mice and the effects of AIMP1 deficiency on production of TH2 cytokines were evaluated in T cells using AIMP1-specific siRNA. Additionally, the enhanced pause values and histologic analysis were assessed in mice receiving AIMP1-deficient CD4+ T cells with OVA challenge. Clinical signs of spontaneous airway inflammation were noted in AIMP1-deficienct mice. AIMP1-deficient mice showed strongly increased Penh values in response to methacholine without any allergen exposure. Adoptive transfer of AIMP1-deficient CD4+ T cells to OVA-sensitized C57BL/6 mice exacerbated OVA-induced airway inflammation and increased infiltration of inflammatory cells into the lung. Furthermore, lung DCs in AIMP1-deficient mice showed increased expression of surface molecules, and IL-12p40 level in sera significantly decreased in AIMP1-deficient mice compared to that of wild type mice. These results strongly indicate that AIMP1 plays a role in negatively regulating TH2 responses in vivo, and AIMP1 can be employed as a novel therapeutic agent against TH2-biased diseases, particularly asthma.
Collapse
|