1
|
Adiga V, Bindhu H, Ahmed A, Chetan Kumar N, Tripathi H, D’Souza G, Dias M, Shivalingaiah S, Rao S, K N S, Hawrylowicz C, Dwarkanath P, Vyakarnam A. Immune profiling reveals umbilical cord blood mononuclear cells from South India display an IL-8 dominant, CXCL-10 deficient polyfunctional monocyte response to pathogen-associated molecular patterns that is distinct from adult blood cells. Clin Exp Immunol 2024; 217:263-278. [PMID: 38695079 PMCID: PMC11310697 DOI: 10.1093/cei/uxae034] [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: 11/30/2023] [Revised: 02/28/2024] [Accepted: 05/14/2024] [Indexed: 08/10/2024] Open
Abstract
Neonate responses to pathogen-associated molecular patterns (PAMPS) differ from adults; such understanding is poor in Indian neonates, despite recognized significant infectious risk. Immune profiling analysis was undertaken of 10 secreted mediators contextualized with cellular source induced by six PAMPs in umbilical cord (CB; n = 21) and adult-blood (PBMC; n = 14) from a tertiary care hospital in South India. Differential cytokine expression analysis (minimum log2-fold difference; adj P-value < 0.05) identified bacterial PAMPs induced higher concentrations of IL-1β, IL-10, TNF-α in adults versus IL-8, GM-CSF, IFN-γ, and IL-2 in CB. CB responded to poly I:C and SARS-CoV-2 lysate with a dominant IL-8 response, whereas in PBMC, CXCL-10 dominated poly I:C, but not SARS-CoV-2, responses, highlighting potential IL-8 importance, in the absence of Type I Interferons, in antiviral CB immunity. Candida albicans was the only PAMP to uniformly induce higher secretion of effectors in CB. The predominant source of IL-8/IL-6/TNF-α/IL-1β in both CB and PBMC was polyfunctional monocytes and IFN-γ/IL-2/IL-17 from innate lymphocytes. Correlation matrix analyses revealed IL-8 to be the most differentially regulated, correlating positively in CB versus negatively in PBMC with IL-6, GM-CSF, IFN-γ, IL-2, consistent with more negatively regulated cytokine modules in adults, potentially linked to higher anti-inflammatory IL-10. Cord and adult blood from India respond robustly to PAMPs with unique effector combinations. These data provide a strong foundation to monitor, explore, mechanisms that regulate such immunity during the life course, an area of significant global health importance given infection-related infant mortality incidence.
Collapse
Affiliation(s)
- Vasista Adiga
- Human Immunology Laboratory, Division of Infectious Diseases, St. John’s Research Institute, Bangalore, Karnataka, India
- Department of Biotechnology, PES University, Bangalore, Karnataka, India
| | - Hima Bindhu
- Human Immunology Laboratory, Division of Infectious Diseases, St. John’s Research Institute, Bangalore, Karnataka, India
| | - Asma Ahmed
- Human Immunology Laboratory, Division of Infectious Diseases, St. John’s Research Institute, Bangalore, Karnataka, India
| | - Nirutha Chetan Kumar
- Human Immunology Laboratory, Division of Infectious Diseases, St. John’s Research Institute, Bangalore, Karnataka, India
| | - Himanshu Tripathi
- Human Immunology Laboratory, Division of Infectious Diseases, St. John’s Research Institute, Bangalore, Karnataka, India
| | - George D’Souza
- Department of Pulmonary Medicine, St. John’s Medical College, Bangalore, India
| | - Mary Dias
- Division of Infectious Diseases, St. John’s Research Institute, Bangalore, Karnataka, India
| | | | - Srishti Rao
- Division of Infectious Diseases, St. John’s Research Institute, Bangalore, Karnataka, India
| | - Shanti K N
- Department of Biotechnology, PES University, Bangalore, Karnataka, India
| | - Catherine Hawrylowicz
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Science & Medicine, King’s College, London, UK
| | - Pratibha Dwarkanath
- Division of Nutrition, St. John’s Research Institute, Bangalore, Karnataka, India
| | - Annapurna Vyakarnam
- Human Immunology Laboratory, Division of Infectious Diseases, St. John’s Research Institute, Bangalore, Karnataka, India
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Science & Medicine, King’s College, London, UK
| |
Collapse
|
2
|
DaMata JP, Zelkoski AE, Nhan PB, Ennis KHE, Kim JS, Lu Z, Malloy AMW. Dissociation protocols influence the phenotypes of lymphocyte and myeloid cell populations isolated from the neonatal lymph node. Front Immunol 2024; 15:1368118. [PMID: 38756770 PMCID: PMC11097666 DOI: 10.3389/fimmu.2024.1368118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/18/2024] [Indexed: 05/18/2024] Open
Abstract
Frequencies and phenotypes of immune cells differ between neonates and adults in association with age-specific immune responses. Lymph nodes (LN) are critical tissue sites to quantify and define these differences. Advances in flow cytometry have enabled more multifaceted measurements of complex immune responses. Tissue processing can affect the immune cells under investigation that influence key findings. To understand the impact on immune cells in the LN after processing for single-cell suspension, we compared three dissociation protocols: enzymatic digestion, mechanical dissociation with DNase I treatment, and mechanical dissociation with density gradient separation. We analyzed cell yields, viability, phenotypic and maturation markers of immune cells from the lung-draining LN of neonatal and adult mice two days after intranasal respiratory syncytial virus (RSV) infection. While viability was consistent across age groups, the protocols influenced the yield of subsets defined by important phenotypic and activation markers. Moreover, enzymatic digestion did not show higher overall yields of conventional dendritic cells and macrophages from the LN. Together, our findings show that the three dissociation protocols have similar impacts on the number and viability of cells isolated from the neonatal and adult LN. However, enzymatic digestion impacts the mean fluorescence intensity of key lineage and activation markers that may influence experimental findings.
Collapse
Affiliation(s)
- Jarina P. DaMata
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Amanda E. Zelkoski
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
| | - Paula B. Nhan
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Katherine H. E. Ennis
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Ji Sung Kim
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Zhongyan Lu
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Allison M. W. Malloy
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
| |
Collapse
|
3
|
Mohamad Zainal NH, Mohd Nor NH, Saat A, Clifton VL. Childhood allergy susceptibility: The role of the immune system development in the in-utero period. Hum Immunol 2022; 83:437-446. [DOI: 10.1016/j.humimm.2022.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 01/20/2022] [Accepted: 02/07/2022] [Indexed: 11/30/2022]
|
4
|
Childhood Allergy Disease, Early Diagnosis, and the Potential of Salivary Protein Biomarkers. Mediators Inflamm 2021; 2021:9198249. [PMID: 34658668 PMCID: PMC8519724 DOI: 10.1155/2021/9198249] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/11/2021] [Indexed: 12/17/2022] Open
Abstract
Allergic disease has risen to epidemic proportions since the last decade and is among the most common noncommunicable, chronic diseases in children and adolescents worldwide. Allergic disease usually occurs in early life; thus, early biomarkers of allergic susceptibility are required for preventive measures to high-risk infants which enable early interventions to decrease allergic severity. However, to date, there is no reliable general or specific allergy phenotype detection method that is easy and noninvasive for children. Most reported allergic phenotype detection methods are invasive, such as the skin prick test (SPT), oral food challenge (OFC), and blood test, and many involve not readily accessible biological samples, such as cord blood (CB), maternal blood, or newborn vernix. Saliva is a biological sample that has great potential as a biomarker measurement as it consists of an abundance of biomarkers, such as genetic material and proteins. It is easily accessible, noninvasive, collected via a painless procedure, and an easy bedside screening for real-time measurement of the ongoing human physiological system. All these advantages emphasise saliva as a very promising diagnostic candidate for the detection and monitoring of disease biomarkers, especially in children. Furthermore, protein biomarkers have the advantages as modifiable influencing factors rather than genetic and epigenetic factors that are mostly nonmodifiable factors for allergic disease susceptibility in childhood. Saliva has great potential to replace serum as a biological fluid biomarker in diagnosing clinical allergy. However, to date, saliva is not considered as an established medically acceptable biomarker. This review considers whether the saliva could be suitable biological samples for early detection of allergic risk. Such tools may be used as justification for targeted interventions in early childhood for disease prevention and assisting in reducing morbidity and mortality caused by childhood allergy.
Collapse
|
5
|
Michael H, Li Y, Wang Y, McCusker CT. Trained immunity induced by in vivo peptide-based STAT6 inhibition prevents ragweed allergy in mice. Allergy Asthma Clin Immunol 2021; 17:42. [PMID: 33883042 PMCID: PMC8059037 DOI: 10.1186/s13223-021-00542-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 04/02/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Trained immunity is the ability of the innate immune system to form immune memory responses to provide support the formation of appropriate adaptive responses. Allergic airways disease (AAD) is a maladapted immune response to allergens, initiated and maintained by the type 2 (T2) inflammatory pathway. It is predicated by the elaboration of cytokines IL-4 and IL-13 and follows activation of the STAT6 transcription factor. OBJECTIVE To investigate the role of trained immunity in mucosal immune responses following neonatal vaccination with the STAT6 inhibitory peptide (STAT6-IP), in preventing the development of ragweed-induced AAD. METHODS We demonstrate that transfer of CD4+ T cells or dendritic cells (DC) from STAT6-IP vaccinated wild-type BALB/c mice to naïve mice, that were subsequently chronically exposed to sensitizing doses of ragweed allergen, is sufficient to prevent development of T2 responses in recipients. RESULTS Our results demonstrate significant reductions in; airways hyperresponsiveness (AHR); ragweed-specific IgE; pulmonary inflammation; T2 cytokines; and inflammatory gene expressions in recipient mice. Expression of IDO, TGFβ and T regulatory cells were all significantly increased. Anti-TGFβ treatment during the ragweed sensitization phase re-constituted the pro-inflammatory T2 immune response. We show that tolerance can be attained via DC trained in the STAT6-IP-mediated tolerant milieu. This effect is not restricted to a particular allergen and does not require antigen-mediated T cell activation prior to transfer. CONCLUSION Adoptive transfer experiments suggest that STAT6-IP treatment trains dendritic and cells to mediate tolerant immunity to chronic ragweed exposure in the airways. This indicates that early transient STAT6-inhibition constitutes an effective immunomodulatory airways allergy preventative strategy.
Collapse
Affiliation(s)
- Husheem Michael
- Meakins-Christie Laboratories, McGill University and the McGill University Health Care-Research Institute, Block E, RI-MUHC, EM3.2219, 1001 Boulevard Décarie, Montréal, QC, H4A 3J1, Canada
| | - Yuanyi Li
- Meakins-Christie Laboratories, McGill University and the McGill University Health Care-Research Institute, Block E, RI-MUHC, EM3.2219, 1001 Boulevard Décarie, Montréal, QC, H4A 3J1, Canada
| | - Yufa Wang
- Meakins-Christie Laboratories, McGill University and the McGill University Health Care-Research Institute, Block E, RI-MUHC, EM3.2219, 1001 Boulevard Décarie, Montréal, QC, H4A 3J1, Canada
| | - Christine T McCusker
- Meakins-Christie Laboratories, McGill University and the McGill University Health Care-Research Institute, Block E, RI-MUHC, EM3.2219, 1001 Boulevard Décarie, Montréal, QC, H4A 3J1, Canada.
| |
Collapse
|
6
|
Abstract
The lung and gut microbiome are factors in asthma risk or protection. Relevant elements of the microbiome within both niches include the importance of the early life window for microbiome establishment, the diversity of bacteria, richness of bacteria, and effect of those bacteria on the local epithelium and immune system. Mechanisms of protection include direct anti-inflammatory action or induction of non-type 2 inflammation by certain bacterial colonies. The gut microbiome further impacts asthma risk through the contribution of metabolic products. This article reviews the mechanisms that connect the lung and gut microbiota to asthma development and severity.
Collapse
Affiliation(s)
- Tara F Carr
- Department of Medicine, University of Arizona, 1501 North Campbell Avenue, Tucson, AZ 85724-5030, USA.
| | - Rhonda Alkatib
- Department of Medicine, University of Arizona, 1501 North Campbell Avenue, Tucson, AZ 85724-5030, USA
| | - Monica Kraft
- Department of Medicine, University of Arizona, 1501 North Campbell Avenue, Tucson, AZ 85724-5030, USA
| |
Collapse
|
7
|
Mincham KT, Scott NM, Lauzon-Joset JF, Leffler J, Larcombe AN, Stumbles PA, Robertson SA, Pasquali C, Holt PG, Strickland DH. Transplacental immune modulation with a bacterial-derived agent protects against allergic airway inflammation. J Clin Invest 2018; 128:4856-4869. [PMID: 30153109 DOI: 10.1172/jci122631] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 08/23/2018] [Indexed: 01/18/2023] Open
Abstract
Chronic allergic inflammatory diseases are a major cause of morbidity, with allergic asthma alone affecting over 300 million people worldwide. Epidemiological studies demonstrate that environmental stimuli are associated with either the promotion or prevention of disease. Major reductions in asthma prevalence are documented in European and US farming communities. Protection is associated with exposure of mothers during pregnancy to microbial breakdown products present in farm dusts and unprocessed foods and enhancement of innate immune competence in the children. We sought to develop a scientific rationale for progressing these findings toward clinical application for primary disease prevention. Treatment of pregnant mice with a defined, clinically approved immune modulator was shown to markedly reduce susceptibility of their offspring to development of the hallmark clinical features of allergic airway inflammatory disease. Mechanistically, offspring displayed enhanced dendritic cell-dependent airway mucosal immune surveillance function, which resulted in more efficient generation of mucosal-homing regulatory T cells in response to local inflammatory challenge. We provide evidence that the principal target for maternal treatment effects was the fetal dendritic cell progenitor compartment, equipping the offspring for accelerated functional maturation of the airway mucosal dendritic cell network following birth. These data provide proof of concept supporting the rationale for developing transplacental immune reprogramming approaches for primary disease prevention.
Collapse
Affiliation(s)
- Kyle T Mincham
- Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
| | - Naomi M Scott
- Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
| | | | - Jonatan Leffler
- Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
| | - Alexander N Larcombe
- Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia.,Health, Safety and Environment, School of Public Health, Curtin University, Perth, Western Australia, Australia
| | - Philip A Stumbles
- Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia.,School of Veterinary and Life Sciences, Murdoch University, Perth, Western Australia, Australia.,School of Paediatrics and Child Health, University of Western Australia, Subiaco, Western Australia, Australia
| | - Sarah A Robertson
- Robinson Research Institute and School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | | | - Patrick G Holt
- Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
| | - Deborah H Strickland
- Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
| |
Collapse
|
8
|
Shattuck EC. Ecological Context and Human Variation: Applying the Principles of Biological Anthropology to Psychoneuroimmunology. Methods Mol Biol 2018; 1781:55-76. [PMID: 29705842 DOI: 10.1007/978-1-4939-7828-1_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
There is considerable research interest overlap between biological anthropology and psychoneuroimmunology (PNI), particularly given recent anthropological interest in endocrine and immune system functioning over the life span and in different environmental contexts. In this chapter, I argue that conducting research on non-WEIRD populations and applying an anthropological, evolutionary approach to PNI can greatly strengthen our understanding of immune-endocrine-behavior connections. This chapter reviews population-level variation in the human immune and endocrine systems, as well as genetic and environmental contributions to this variation. The effects of culture on shaping health outcomes and stress responses are also considered. Finally, this chapter discusses some noninvasive sampling methodologies appropriate to field research and alternatives to laboratory-based research designs. By confronting variable social and environmental contexts, PNI can greatly expand on its existing contributions to the treatment and understanding of depression, mood disorders, stress, and other aspects of health and well-being.
Collapse
Affiliation(s)
- Eric C Shattuck
- Department of Anthropology, University of Texas at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
9
|
Cord blood transplantation recapitulates fetal ontogeny with a distinct molecular signature that supports CD4 + T-cell reconstitution. Blood Adv 2017; 1:2206-2216. [PMID: 29296868 DOI: 10.1182/bloodadvances.2017010827] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 09/16/2017] [Indexed: 12/27/2022] Open
Abstract
Omission of in vivo T-cell depletion promotes rapid, thymic-independent CD4+-biased T-cell recovery after cord blood transplant. This enhanced T-cell reconstitution differs from that seen after stem cell transplant from other stem cell sources, but the mechanism is not known. Here, we demonstrate that the transcription profile of naive CD4+ T cells from cord blood and that of lymphocytes reconstituting after cord blood transplantation is similar to the transcription profile of fetal CD4+ T cells. This profile is distinct to that of naive CD4+ T cells from peripheral blood and that of lymphocytes reconstituting after T-replete bone marrow transplantation. The transcription profile of reconstituting naive CD4+ T cells from cord blood transplant recipients was upregulated in the T-cell receptor (TCR) signaling pathway and its transcription factor activator protein-1 (AP-1). Furthermore, a small molecule inhibitor of AP-1 proportionally inhibited cord blood CD4+ T-cell proliferation (P < .05). Together, these findings suggest that reconstituting cord blood CD4+ T cells reflect the properties of fetal ontogenesis, and enhanced TCR signaling is responsible for the rapid restoration of the unique CD4+ T-cell biased adaptive immunity after cord blood transplantation.
Collapse
|
10
|
Súkeníková L, Černý V, Novotná O, Petrásková P, Boráková K, Kolářová L, Prokešová L, Hrdý J. Different capacity of in vitro generated myeloid dendritic cells of newborns of healthy and allergic mothers to respond to probiotic strain E. coli O83:K24:H31. Immunol Lett 2017; 189:82-89. [PMID: 28554713 DOI: 10.1016/j.imlet.2017.05.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 05/19/2017] [Accepted: 05/23/2017] [Indexed: 10/19/2022]
Abstract
Allergic diseases belong to one of the most common diseases with steadily increasing incidence even among young children. There is an urgent need to identify a prognostic marker pointing to increased risk of allergy development enabling early preventive measures introduction. It has been shown that administration of selected probiotic strains or mixtures could prevent allergy development. In our study, we have tested the capacity of probiotic strain Escherichia coli O83:K24:H31 (E. coli O83) to promote dendritic cell (DC) maturation and polarisation of immune responses. Increased presence of activation marker CD83 was observed on DC stimulated by E. coli O83 and DC of newborns of allergic mothers have significantly more increased cell surface presence of CD83 in comparison to children of healthy mothers. Increased gene expression and secretion of IL-10 was detected in DC stimulated with E. coli O83 being higher in DC of newborns of healthy mothers in comparison to allergic ones. Generally, increased presence of intracellular cytokines (IL-4, IL-13, IFN-gamma, IL-17A, IL-22, IL-10) was detected in CD4+ T cells cocultured with DC of children of allergic mothers in comparison to healthy ones. E. coli O83 primed DC significantly increased IL-10 and IL-17A in CD4 T cells of newborns of healthy mothers in comparison to the levels detected in CD4 T cells cocultured with control non-stimulated DC. We can conclude E. coli O83 induces dendritic cell maturation and IL-10 production in DC. Newborns of allergic mothers have generally increased reactivity of both DC and CD4 T cells which together with decreased capacity of DC of newborns of allergic mothers to produce IL-10 could support inappropriate immune responses development after allergen encounter.
Collapse
Affiliation(s)
- Lenka Súkeníková
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Viktor Černý
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Olga Novotná
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Petra Petrásková
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | | | - Libuše Kolářová
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Ludmila Prokešová
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Jiří Hrdý
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic.
| |
Collapse
|
11
|
Renz H, Holt PG, Inouye M, Logan AC, Prescott SL, Sly PD. An exposome perspective: Early-life events and immune development in a changing world. J Allergy Clin Immunol 2017; 140:24-40. [DOI: 10.1016/j.jaci.2017.05.015] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 05/22/2017] [Accepted: 05/23/2017] [Indexed: 02/09/2023]
|
12
|
The infant gut bacterial microbiota and risk of pediatric asthma and allergic diseases. Transl Res 2017; 179:60-70. [PMID: 27469270 PMCID: PMC5555614 DOI: 10.1016/j.trsl.2016.06.010] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 06/26/2016] [Accepted: 06/30/2016] [Indexed: 02/07/2023]
Abstract
Among the many areas being revolutionized by the recent introduction of culture-independent microbial identification techniques is investigation of the relationship between close contact with large animals, antibiotics, breast feeding, mode of birth, and other exposures during infancy as related to a reduced risk of asthma and allergic disease. These exposures were originally clustered under the "Hygiene Hypothesis" which has evolved into the "Microbiota Hypothesis". This review begins by summarizing epidemiologic studies suggesting that the common feature of these allergy risk-related exposures is their influence on the founding and early development of a child's gut microbiota. Next, studies using culture-independent techniques are presented showing that children who have experienced the exposures of interest have altered gut microbiota. Finally, selected mouse and human studies are presented which begin to corroborate the protective exposures identified in epidemiologic studies by elucidating mechanisms through which microbes can alter immune development and function. These microbially driven immune alterations demonstrate that microbial exposures in many cases could alter the risk of subsequent allergic disease and asthma. Hopefully, a better understanding of how microbes influence allergic disease will lead to safe and effective methods for reducing the prevalence of all forms of allergic disease.
Collapse
|
13
|
Francis JP, Richmond PC, Strickland D, Prescott SL, Pomat WS, Michael A, Nadal-Sims MA, Edwards-Devitt CJ, Holt PG, Lehmann D, van den Biggelaar AHJ. Cord blood Streptococcus pneumoniae-specific cellular immune responses predict early pneumococcal carriage in high-risk infants in Papua New Guinea. Clin Exp Immunol 2016; 187:408-417. [PMID: 27859014 PMCID: PMC5290304 DOI: 10.1111/cei.12902] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 10/03/2016] [Accepted: 10/27/2016] [Indexed: 12/01/2022] Open
Abstract
In areas where Streptococcus pneumoniae is highly endemic, infants experience very early pneumococcal colonization of the upper respiratory tract, with carriage often persisting into adulthood. We aimed to explore whether newborns in high‐risk areas have pre‐existing pneumococcal‐specific cellular immune responses that may affect early pneumococcal acquisition. Cord blood mononuclear cells (CBMC) of 84 Papua New Guinean (PNG; high endemic) and 33 Australian (AUS; low endemic) newborns were stimulated in vitro with detoxified pneumolysin (dPly) or pneumococcal surface protein A (PspA; families 1 and 2) and compared for cytokine responses. Within the PNG cohort, associations between CBMC dPly and PspA‐induced responses and pneumococcal colonization within the first month of life were studied. Significantly higher PspA‐specific interferon (IFN)‐γ, tumour necrosis factor (TNF)‐α, interleukin (IL)‐5, IL‐6, IL‐10 and IL‐13 responses, and lower dPly‐IL‐6 responses were produced in CBMC cultures of PNG compared to AUS newborns. Higher CBMC PspA‐IL‐5 and PspA‐IL‐13 responses correlated with a higher proportion of cord CD4 T cells, and higher dPly‐IL‐6 responses with a higher frequency of cord antigen‐presenting cells. In the PNG cohort, higher PspA‐specific IL‐5 and IL‐6 CBMC responses were associated independently and significantly with increased risk of earlier pneumococcal colonization, while a significant protective effect was found for higher PspA‐IL‐10 CBMC responses. Pneumococcus‐specific cellular immune responses differ between children born in pneumococcal high versus low endemic settings, which may contribute to the higher risk of infants in high endemic settings for early pneumococcal colonization, and hence disease.
Collapse
Affiliation(s)
- J P Francis
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
| | - P C Richmond
- School of Paediatrics and Child Health, University of Western Australia, Perth, Australia
| | - D Strickland
- Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - S L Prescott
- School of Paediatrics and Child Health, University of Western Australia, Perth, Australia
| | - W S Pomat
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
| | - A Michael
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
| | - M A Nadal-Sims
- Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - C J Edwards-Devitt
- Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - P G Holt
- Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - D Lehmann
- Telethon Kids Institute, University of Western Australia, Perth, Australia
| | | |
Collapse
|
14
|
Morris MC, Surendran N. Neonatal Vaccination: Challenges and Intervention Strategies. Neonatology 2016; 109:161-9. [PMID: 26757146 PMCID: PMC4749499 DOI: 10.1159/000442460] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Accepted: 11/16/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND While vaccines have been tremendously successful in reducing the incidence of serious infectious diseases, newborns remain particularly vulnerable in the first few months of their life to life-threatening infections. A number of challenges exist to neonatal vaccination. However, recent advances in the understanding of neonatal immunology offer insights to overcome many of those challenges. OBJECTIVE This review will present an overview of the features of neonatal immunity which make vaccination difficult, survey the mechanisms of action of available vaccine adjuvants with respect to the unique features of neonatal immunity, and propose a possible mechanism contributing to the inability of neonates to generate protective immune responses to vaccines. METHODS We surveyed recent published findings on the challenges to neonatal vaccination and possible intervention strategies including the use of novel vaccine adjuvants to develop efficacious neonatal vaccines. RESULTS Challenges in the vaccination of neonates include interference from maternal antibody and excessive skewing towards Th2 immunity, which can be counteracted by the use of proper adjuvants. CONCLUSION Synergistic stimulation of multiple Toll-like receptors by incorporating well-defined agonist-adjuvant combinations to vaccines is a promising strategy to ensure a protective vaccine response in neonates.
Collapse
Affiliation(s)
- Matthew C Morris
- Research Institute, Rochester Regional Health Systems, Rochester, N.Y., USA
| | | |
Collapse
|
15
|
TGF-β-mediated airway tolerance to allergens induced by peptide-based immunomodulatory mucosal vaccination. Mucosal Immunol 2015; 8:1248-61. [PMID: 25783968 DOI: 10.1038/mi.2015.15] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 01/30/2015] [Indexed: 02/07/2023]
Abstract
We sought to modulate mucosal immune responses using neonatal vaccination to avert the development of allergic airways disease (AAD). Pulmonary pathology in AAD is driven by T helper (TH)2 cytokines, in particular interleukin (IL)4 and IL13, the expression and actions of which are regulated by the transcription factor STAT6. We developed a peptide homolog of STAT6, STAT6-IP. Neonatal mice given, intranasally, STAT6-IP, in an effort to modulate de novo airways immune responses, developed tolerance following subsequent allergen sensitization, with either ovalbumin or ragweed allergens, as demonstrated by reduced TH2 cytokines and specific immunoglobulin (Ig)E and the significant increases in the latency-associated peptide (LAP)(+) T-regulatory (Treg) cell subset and expression of transforming growth factor (TGF)-β. This regulatory phenotype was transferrable by CD4(+) T cells or CD11c(+) dendritic cells (DCs) derived from STAT6-IP-vaccinated mice. Anti-TGF-β treatment during allergen sensitization, however, re-established the pro-inflammatory TH2 response. Thus, neonatal STAT6-IP vaccination induces prospective TGF-β-dependent tolerance to allergen and constitutes a novel highly effective immunomodulatory allergy prevention strategy.
Collapse
|
16
|
Bousquet J, Anto JM, Wickman M, Keil T, Valenta R, Haahtela T, Lodrup Carlsen K, van Hage M, Akdis C, Bachert C, Akdis M, Auffray C, Annesi-Maesano I, Bindslev-Jensen C, Cambon-Thomsen A, Carlsen KH, Chatzi L, Forastiere F, Garcia-Aymerich J, Gehrig U, Guerra S, Heinrich J, Koppelman GH, Kowalski ML, Lambrecht B, Lupinek C, Maier D, Melén E, Momas I, Palkonen S, Pinart M, Postma D, Siroux V, Smit HA, Sunyer J, Wright J, Zuberbier T, Arshad SH, Nadif R, Thijs C, Andersson N, Asarnoj A, Ballardini N, Ballereau S, Bedbrook A, Benet M, Bergstrom A, Brunekreef B, Burte E, Calderon M, De Carlo G, Demoly P, Eller E, Fantini MP, Hammad H, Hohman C, Just J, Kerkhof M, Kogevinas M, Kull I, Lau S, Lemonnier N, Mommers M, Nawijn M, Neubauer A, Oddie S, Pellet J, Pin I, Porta D, Saes Y, Skrindo I, Tischer CG, Torrent M, von Hertzen L. Are allergic multimorbidities and IgE polysensitization associated with the persistence or re-occurrence of foetal type 2 signalling? The MeDALL hypothesis. Allergy 2015; 70:1062-78. [PMID: 25913421 DOI: 10.1111/all.12637] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2015] [Indexed: 12/22/2022]
Abstract
Allergic diseases [asthma, rhinitis and atopic dermatitis (AD)] are complex. They are associated with allergen-specific IgE and nonallergic mechanisms that may coexist in the same patient. In addition, these diseases tend to cluster and patients present concomitant or consecutive diseases (multimorbidity). IgE sensitization should be considered as a quantitative trait. Important clinical and immunological differences exist between mono- and polysensitized subjects. Multimorbidities of allergic diseases share common causal mechanisms that are only partly IgE-mediated. Persistence of allergic diseases over time is associated with multimorbidity and/or IgE polysensitization. The importance of the family history of allergy may decrease with age. This review puts forward the hypothesis that allergic multimorbidities and IgE polysensitization are associated and related to the persistence or re-occurrence of foetal type 2 signalling. Asthma, rhinitis and AD are manifestations of a common systemic immune imbalance (mesodermal origin) with specific patterns of remodelling (ectodermal or endodermal origin). This study proposes a new classification of IgE-mediated allergic diseases that allows the definition of novel phenotypes to (i) better understand genetic and epigenetic mechanisms, (ii) better stratify allergic preschool children for prognosis and (iii) propose novel strategies of treatment and prevention.
Collapse
Affiliation(s)
- J. Bousquet
- University Hospital; Montpellier France
- MACVIA-LR; Contre les MAladies Chroniques pour un VIeillissement Actif en Languedoc-Roussillon; European Innovation Partnership on Active and Healthy Ageing Reference Site; Paris France
- INSERM; VIMA: Ageing and Chronic Diseases Epidemiological and Public Health Approaches, U1168; Paris France
- UVSQ; UMR-S 1168; Université Versailles St-Quentin-en-Yvelines; Versailles France
| | - J. M. Anto
- Centre for Research in Environmental Epidemiology (CREAL); Barcelona Spain
- Hospital del Mar Research Institute (IMIM); Barcelona Spain
- CIBER Epidemiología y Salud Pública (CIBERESP); Barcelona Spain
- Department of Experimental and Health Sciences; University of Pompeu Fabra (UPF); Barcelona Spain
| | - M. Wickman
- Sachs’ Children's Hospital; Stockholm Sweden
- Institute of Environmental Medicine; Karolinska Institutet; Stockholm Sweden
| | - T. Keil
- Institute of Social Medicine, Epidemiology and Health Economics; Charité - Universitätsmedizin Berlin; Berlin Germany
- Institute for Clinical Epidemiology and Biometry; University of Wuerzburg; Wuerzburg Germany
| | - R. Valenta
- Division of Immunopathology; Department of Pathophysiology and Allergy Research; Center for Pathophysiology, Infectiology and Immunology; Medical University of Vienna; Vienna Austria
| | - T. Haahtela
- Skin and Allergy Hospital; Helsinki University Hospital; Helsinki Finland
| | - K. Lodrup Carlsen
- Department of Paediatrics; Oslo University Hospital; Oslo Norway
- Faculty of Medicine; Institute of Clinical Medicine; University of Oslo; Oslo Norway
| | - M. van Hage
- Clinical Immunology and Allergy Unit; Department of Medicine Solna; Karolinska Institutet and University Hospital; Stockholm Sweden
| | - C. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zurich; Davos Switzerland
| | - C. Bachert
- ENT Department; Ghent University Hospital; Gent Belgium
| | - M. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zurich; Davos Switzerland
| | - C. Auffray
- European Institute for Systems Biology and Medicine; Lyon France
| | - I. Annesi-Maesano
- EPAR U707 INSERM; Paris France
- EPAR UMR-S UPMC; Paris VI; Paris France
| | - C. Bindslev-Jensen
- Department of Dermatology and Allergy Centre; Odense University Hospital; Odense Denmark
| | - A. Cambon-Thomsen
- UMR Inserm U1027; Université de Toulouse III Paul Sabatier; Toulouse France
| | - K. H. Carlsen
- Department of Paediatrics; Oslo University Hospital; Oslo Norway
- University of Oslo; Oslo Norway
| | - L. Chatzi
- Department of Social Medicine; Faculty of Medicine; University of Crete; Heraklion Crete Greece
| | - F. Forastiere
- Department of Epidemiology; Regional Health Service Lazio Region; Rome Italy
| | - J. Garcia-Aymerich
- Centre for Research in Environmental Epidemiology (CREAL); Barcelona Spain
- Hospital del Mar Research Institute (IMIM); Barcelona Spain
- CIBER Epidemiología y Salud Pública (CIBERESP); Barcelona Spain
- Department of Experimental and Health Sciences; University of Pompeu Fabra (UPF); Barcelona Spain
| | - U. Gehrig
- Julius Center of Health Sciences and Primary Care; University Medical Center Utrecht; University of Utrecht; Utrecht the Netherlands
| | - S. Guerra
- Centre for Research in Environmental Epidemiology (CREAL); Barcelona Spain
| | - J. Heinrich
- Institute of Epidemiology; German Research Centre for Environmental Health; Helmholtz Zentrum München; Neuherberg Germany
| | - G. H. Koppelman
- Department of Pediatric Pulmonology and Pediatric Allergology; GRIAC Research Institute; University Medical Center Groningen; Beatrix Children's Hospital; University of Groningen; Groningen the Netherlands
| | - M. L. Kowalski
- Department of Immunology, Rheumatology and Allergy; Medical University of Lodz; Lodz Poland
| | - B. Lambrecht
- VIB Inflammation Research Center; Ghent University; Ghent Belgium
| | - C. Lupinek
- Division of Immunopathology; Department of Pathophysiology and Allergy Research; Center for Pathophysiology, Infectiology and Immunology; Medical University of Vienna; Vienna Austria
| | | | - E. Melén
- Institute of Environmental Medicine; Karolinska Institutet; Stockholm Sweden
| | - I. Momas
- Department of Public Health and Biostatistics, EA 4064; Paris Descartes University; Paris France
- Paris Municipal Department of Social Action, Childhood, and Health; Paris France
| | - S. Palkonen
- EFA European Federation of Allergy and Airways Diseases Patients' Associations; Brussels Belgium
| | - M. Pinart
- Centre for Research in Environmental Epidemiology (CREAL); Barcelona Spain
| | - D. Postma
- Department of Respiratory Medicine; GRIAC Research Institute; University Medical Center Groningen; Beatrix Children's Hospital; University of Groningen; Groningen the Netherlands
| | | | - H. A. Smit
- Julius Center of Health Sciences and Primary Care; University Medical Center Utrecht; University of Utrecht; Utrecht the Netherlands
| | - J. Sunyer
- Centre for Research in Environmental Epidemiology (CREAL); Barcelona Spain
- Hospital del Mar Research Institute (IMIM); Barcelona Spain
- CIBER Epidemiología y Salud Pública (CIBERESP); Barcelona Spain
- Department of Experimental and Health Sciences; University of Pompeu Fabra (UPF); Barcelona Spain
| | - J. Wright
- Bradford Institute for Health Research; Bradford Royal Infirmary; Bradford UK
| | - T. Zuberbier
- Allergy-Centre-Charité at the Department of Dermatology; Charité - Universitätsmedizin Berlin; Berlin Germany
- Secretary General of the Global Allergy and Asthma European Network (GA2LEN); Berlin Germany
| | - S. H. Arshad
- David Hide Asthma and Allergy Research Centre; Isle of Wight UK
| | - R. Nadif
- INSERM; VIMA: Ageing and Chronic Diseases Epidemiological and Public Health Approaches, U1168; Paris France
- UVSQ; UMR-S 1168; Université Versailles St-Quentin-en-Yvelines; Versailles France
| | - C. Thijs
- Department of Epidemiology; CAPHRI School of Public Health and Primary Care; Maastricht University; Maastricht the Netherlands
| | - N. Andersson
- Sachs’ Children's Hospital; Stockholm Sweden
- Institute of Environmental Medicine; Karolinska Institutet; Stockholm Sweden
| | - A. Asarnoj
- Sachs’ Children's Hospital; Stockholm Sweden
- Institute of Environmental Medicine; Karolinska Institutet; Stockholm Sweden
| | - N. Ballardini
- Sachs’ Children's Hospital; Stockholm Sweden
- Institute of Environmental Medicine; Karolinska Institutet; Stockholm Sweden
| | - S. Ballereau
- European Institute for Systems Biology and Medicine; Lyon France
| | - A. Bedbrook
- MACVIA-LR; Contre les MAladies Chroniques pour un VIeillissement Actif en Languedoc-Roussillon; European Innovation Partnership on Active and Healthy Ageing Reference Site; Paris France
| | - M. Benet
- Centre for Research in Environmental Epidemiology (CREAL); Barcelona Spain
| | - A. Bergstrom
- Sachs’ Children's Hospital; Stockholm Sweden
- Institute of Environmental Medicine; Karolinska Institutet; Stockholm Sweden
| | - B. Brunekreef
- Julius Center of Health Sciences and Primary Care; University Medical Center Utrecht; University of Utrecht; Utrecht the Netherlands
| | - E. Burte
- INSERM; VIMA: Ageing and Chronic Diseases Epidemiological and Public Health Approaches, U1168; Paris France
- UVSQ; UMR-S 1168; Université Versailles St-Quentin-en-Yvelines; Versailles France
| | - M. Calderon
- National Heart and Lung Institute; Imperial College London; Royal Brompton Hospital NHS; London UK
| | - G. De Carlo
- EFA European Federation of Allergy and Airways Diseases Patients' Associations; Brussels Belgium
| | - P. Demoly
- Department of Respiratory Diseases; Montpellier University Hospital; Montpellier France
| | - E. Eller
- Department of Dermatology and Allergy Centre; Odense University Hospital; Odense Denmark
| | - M. P. Fantini
- Department of Medicine and Public Health; Alma Mater Studiorum - University of Bologna; Bologna Italy
| | - H. Hammad
- VIB Inflammation Research Center; Ghent University; Ghent Belgium
| | - C. Hohman
- Institute of Social Medicine, Epidemiology and Health Economics; Charité - Universitätsmedizin Berlin; Berlin Germany
| | - J. Just
- Allergology Department; Centre de l'Asthme et des Allergies; Hôpital d'Enfants Armand-Trousseau (APHP); Paris France
- Institut Pierre Louis d'Epidémiologie et de Santé Publique; Equipe EPAR; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 1136; Paris France
| | - M. Kerkhof
- Department of Respiratory Medicine; GRIAC Research Institute; University Medical Center Groningen; Beatrix Children's Hospital; University of Groningen; Groningen the Netherlands
| | - M. Kogevinas
- Centre for Research in Environmental Epidemiology (CREAL); Barcelona Spain
- Hospital del Mar Research Institute (IMIM); Barcelona Spain
- CIBER Epidemiología y Salud Pública (CIBERESP); Barcelona Spain
- Department of Experimental and Health Sciences; University of Pompeu Fabra (UPF); Barcelona Spain
| | - I. Kull
- Sachs’ Children's Hospital; Stockholm Sweden
- Institute of Environmental Medicine; Karolinska Institutet; Stockholm Sweden
| | - S. Lau
- Department for Pediatric Pneumology and Immunology; Charité Medical University; Berlin Germany
| | - N. Lemonnier
- European Institute for Systems Biology and Medicine; Lyon France
| | - M. Mommers
- Department of Epidemiology; CAPHRI School of Public Health and Primary Care; Maastricht University; Maastricht the Netherlands
| | - M. Nawijn
- Department of Pediatric Pulmonology and Pediatric Allergology; GRIAC Research Institute; University Medical Center Groningen; Beatrix Children's Hospital; University of Groningen; Groningen the Netherlands
| | | | - S. Oddie
- Bradford Institute for Health Research; Bradford Royal Infirmary; Bradford UK
| | - J. Pellet
- European Institute for Systems Biology and Medicine; Lyon France
| | - I. Pin
- Département de pédiatrie; CHU de Grenoble; Grenoble Cedex 9 France
| | - D. Porta
- Department of Epidemiology; Regional Health Service Lazio Region; Rome Italy
| | - Y. Saes
- VIB Inflammation Research Center; Ghent University; Ghent Belgium
| | - I. Skrindo
- Department of Paediatrics; Oslo University Hospital; Oslo Norway
- Faculty of Medicine; Institute of Clinical Medicine; University of Oslo; Oslo Norway
| | - C. G. Tischer
- Institute of Epidemiology; German Research Centre for Environmental Health; Helmholtz Zentrum München; Neuherberg Germany
| | - M. Torrent
- Centre for Research in Environmental Epidemiology (CREAL); Barcelona Spain
- Area de Salut de Menorca, ib-salut; Illes Balears Spain
| | - L. von Hertzen
- Skin and Allergy Hospital; Helsinki University Hospital; Helsinki Finland
| |
Collapse
|
17
|
West CE, Rydén P, Lundin D, Engstrand L, Tulic MK, Prescott SL. Gut microbiome and innate immune response patterns in IgE-associated eczema. Clin Exp Allergy 2015; 45:1419-29. [DOI: 10.1111/cea.12566] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 04/01/2015] [Accepted: 04/29/2015] [Indexed: 02/06/2023]
Affiliation(s)
- C. E. West
- International Inflammation Network (in-FLAME) of the World Universities Network; Umeå Sweden
- Department of Clinical Sciences, Pediatrics; Umeå University; Umeå Sweden
| | - P. Rydén
- Department of Mathematics and Mathematical Statistics; Umeå University; Umeå Sweden
| | - D. Lundin
- Department of Microbiology, Tumor and Cell Biology; Science for Life Laboratory; Karolinska Institutet; Stockholm Sweden
- Bioinformatics Infrastructure for Life Sciences; Science for Life Laboratory; Stockholm Sweden
| | - L. Engstrand
- Department of Microbiology, Tumor and Cell Biology; Science for Life Laboratory; Karolinska Institutet; Stockholm Sweden
| | - M. K. Tulic
- International Inflammation Network (in-FLAME) of the World Universities Network; Umeå Sweden
- Université de Nice Sophia-Antipolis; Nice France
| | - S. L. Prescott
- International Inflammation Network (in-FLAME) of the World Universities Network; Umeå Sweden
- School of Paediatrics and Child Health; University of Western Australia; Perth Australia
| |
Collapse
|
18
|
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
|
19
|
Abstract
The immune system in early life goes through rapid and radical changes. Early life is also the period with the highest risk of infections. The foetal immune system is programmed to coexist with foreign antigenic influences in utero, and postnatally to rapidly develop a functional system capable of distinguishing helpful microbes from harmful pathogens. Both host genetics and environmental influences shape this dramatic transition and direct the trajectory of the developing immune system into early childhood and beyond. Given the malleability of the immune system in early life, interventions aimed at modulating this trajectory thus have the potential to translate into considerable reductions in infectious disease burden with immediate as well as long-lasting benefit. However, an improved understanding of the underlying molecular drivers of early life immunity is prerequisite to optimise such interventions and transform the window of early life vulnerability into one of opportunity.
Collapse
|
20
|
Campbell DE, Boyle RJ, Thornton CA, Prescott SL. Mechanisms of allergic disease - environmental and genetic determinants for the development of allergy. Clin Exp Allergy 2015; 45:844-858. [DOI: 10.1111/cea.12531] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- D. E. Campbell
- Children's Hospital Westmead; Sydney NSW Australia
- Discipline of Paediatrics and Child Health; University of Sydney; Sydney NSW Australia
| | - R. J. Boyle
- Section of Paediatrics; Faculty of Medicine; Imperial College; London UK
| | - C. A. Thornton
- Institute of Life Science; College of Medicine; Swansea University; Swansea UK
| | - S. L. Prescott
- School of Paediatrics and Child Health and Telethon KIDS Institute; c/o Princess Margaret Hospital; University of Western Australia; Perth WA Australia
| |
Collapse
|
21
|
Kallionpää H, Laajala E, Öling V, Härkönen T, Tillmann V, Dorshakova NV, Ilonen J, Lähdesmäki H, Knip M, Lahesmaa R. Standard of hygiene and immune adaptation in newborn infants. Clin Immunol 2014; 155:136-147. [PMID: 25245264 DOI: 10.1016/j.clim.2014.09.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 09/08/2014] [Accepted: 09/10/2014] [Indexed: 01/09/2023]
Abstract
The prevalence of immune-mediated diseases, such as allergies and type 1 diabetes, is on the rise in the developed world. In order to explore differences in the gene expression patterns induced in utero in infants born in contrasting standards of living and hygiene, we collected umbilical cord blood RNA samples from infants born in Finland (modern society), Estonia (rapidly developing society) and the Republic of Karelia, Russia (poor economic conditions). The whole blood transcriptome of Finnish and Estonian neonates differed from their Karelian counterparts, suggesting exposure to toll-like receptor (TLR) ligands and a more matured immune response in infants born in Karelia. These results further support the concept of a conspicuous plasticity in the developing immune system: the environmental factors that play a role in the susceptibility/protection towards immune-mediated diseases begin to shape the neonatal immunity already in utero and direct the maturation in accordance with the surrounding microbial milieu.
Collapse
Affiliation(s)
- Henna Kallionpää
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20521 Turku, Finland; Turku Doctoral Programme of Biomedical Sciences, Kiinamyllynkatu 13, 20520 Turku, Finland; The Finnish Centre of Excellence in Molecular Systems Immunology and Physiology Research, Finland
| | - Essi Laajala
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20521 Turku, Finland; The Finnish Centre of Excellence in Molecular Systems Immunology and Physiology Research, Finland; The National Graduate School in Informational and Structural Biology, 20520 Turku, Finland; Department of Information and Computer Science, Aalto University School of Science, 00076 Aalto, Finland
| | - Viveka Öling
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20521 Turku, Finland; The Finnish Centre of Excellence in Molecular Systems Immunology and Physiology Research, Finland
| | - Taina Härkönen
- The Finnish Centre of Excellence in Molecular Systems Immunology and Physiology Research, Finland; Children's Hospital, University of Helsinki and Helsinki University Central Hospital, 00014 Helsinki, Finland; Diabetes and Obesity Research Program, University of Helsinki, 00014 Helsinki, Finland
| | - Vallo Tillmann
- Department of Pediatrics, University of Tartu and Tartu University Hospital, 51014 Tartu, Estonia
| | | | - Jorma Ilonen
- Immunogenetics Laboratory, University of Turku, 20520 Turku, Finland; Department of Clinical Microbiology, University of Eastern Finland, 70210 Kuopio, Finland
| | - Harri Lähdesmäki
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20521 Turku, Finland; The Finnish Centre of Excellence in Molecular Systems Immunology and Physiology Research, Finland; Department of Information and Computer Science, Aalto University School of Science, 00076 Aalto, Finland
| | - Mikael Knip
- The Finnish Centre of Excellence in Molecular Systems Immunology and Physiology Research, Finland; Children's Hospital, University of Helsinki and Helsinki University Central Hospital, 00014 Helsinki, Finland; Diabetes and Obesity Research Program, University of Helsinki, 00014 Helsinki, Finland; Folkhälsan Research Center, 00290 Helsinki, Finland; Department of Pediatrics, Tampere University Hospital, 33521 Tampere, Finland
| | - Riitta Lahesmaa
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20521 Turku, Finland; The Finnish Centre of Excellence in Molecular Systems Immunology and Physiology Research, Finland.
| | | |
Collapse
|
22
|
Smolen KK, Cai B, Fortuno ES, Gelinas L, Larsen M, Speert DP, Chamekh M, Cooper PJ, Esser M, Marchant A, Kollmann TR. Single-cell analysis of innate cytokine responses to pattern recognition receptor stimulation in children across four continents. THE JOURNAL OF IMMUNOLOGY 2014; 193:3003-3012. [PMID: 25135829 DOI: 10.4049/jimmunol.1400895] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Innate immunity instructs adaptive immunity, and suppression of innate immunity is associated with an increased risk for infection. We showed previously that whole-blood cellular components from a cohort of South African children secreted significantly lower levels of most cytokines following stimulation of pattern recognition receptors compared with whole blood from cohorts of Ecuadorian, Belgian, or Canadian children. To begin dissecting the responsible molecular mechanisms, we set out to identify the relevant cellular source of these differences. Across the four cohorts represented in our study, we identified significant variation in the cellular composition of whole blood; however, a significant reduction in the intracellular cytokine production on the single-cell level was only detected in South African children's monocytes, conventional dendritic cells, and plasmacytoid dendritic cells. We also uncovered a marked reduction in polyfunctionality for each of these cellular compartments in South African children compared with children from the other continents. Together, our data identify differences in cell composition, as well as profoundly lower functional responses of innate cells, in our cohort of South African children. A possible link between altered innate immunity and increased risk for infection or lower response to vaccines in South African infants needs to be explored.
Collapse
Affiliation(s)
- Kinga K Smolen
- Department of Experimental Medicine, University of British Columbia, CFRI A5-147, 950 W28th Ave, Vancouver, BC, V5Z 4H4 Canada
| | - Bing Cai
- Department of Pediatrics, University of British Columbia, CFRI A5-147, 950 W28th Ave, Vancouver, BC, V5Z 4H4 Canada
| | - Edgardo S Fortuno
- Department of Pediatrics, University of British Columbia, CFRI A5-147, 950 W28th Ave, Vancouver, BC, V5Z 4H4 Canada
| | - Laura Gelinas
- Department of Experimental Medicine, University of British Columbia, CFRI A5-147, 950 W28th Ave, Vancouver, BC, V5Z 4H4 Canada
| | - Martin Larsen
- INSERM, U1135, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), F-75013, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie, Paris 06, CR7, CIMI-Paris, F-75013, Paris, France
| | - David P Speert
- Department of Pediatrics, University of British Columbia, CFRI A5-147, 950 W28th Ave, Vancouver, BC, V5Z 4H4 Canada
| | - Mustapha Chamekh
- Institut d'Immunologie, Université Libre de Bruxelles, Charleroi, Rue Adrienne Bolland 8, Gosselies B-6041, Belgium
| | - Philip J Cooper
- Centro de Investigaciones FEPIS, Esmeraldas Quininde, Ecuador, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK.,Molecular and Biochemical Parasitology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK.,Centro de Investgación en Enfermedades Infecciosas, Escuela de Biología, Pontificia Universidad Católica del Ecuador, Casilla 17-22-20418, Quito, Ecuador Ecuador
| | - Monika Esser
- Immunology Unit, Division of Medical Microbiology, Department of Pathology, National Health Laboratory Services and Stellenbosch University, Private Bag X1, Matieland, 7602 South Africa
| | - Arnaud Marchant
- Institut d'Immunologie, Université Libre de Bruxelles, Charleroi, Rue Adrienne Bolland 8, Gosselies B-6041, Belgium.,Département de pédiatrie, Centre Hospitalier Universitaire Saint-Pierre, Rue Haute, 322 1000 Brussels, Belgium
| | - Tobias R Kollmann
- Department of Experimental Medicine, University of British Columbia, CFRI A5-147, 950 W28th Ave, Vancouver, BC, V5Z 4H4 Canada.,Department of Pediatrics, University of British Columbia, CFRI A5-147, 950 W28th Ave, Vancouver, BC, V5Z 4H4 Canada
| |
Collapse
|
23
|
Pizzutto SJ, Yerkovich ST, Upham JW, Hales BJ, Thomas WR, Chang AB. Children with chronic suppurative lung disease have a reduced capacity to synthesize interferon-gamma in vitro in response to non-typeable Haemophilus influenzae. PLoS One 2014; 9:e104236. [PMID: 25111142 PMCID: PMC4128648 DOI: 10.1371/journal.pone.0104236] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 07/08/2014] [Indexed: 01/01/2023] Open
Abstract
Chronic suppurative lung disease (CSLD) is characterized by the presence of a chronic wet or productive cough and recurrent lower respiratory infections. The aim of this study was to identify features of innate, cell-mediated and humoral immunity that may increase susceptibility to respiratory infections in children with CSLD. Because non-typeable Haemophilus influenzae (NTHi) is commonly isolated from the airways in CSLD, we examined immune responses to this organism in 80 age-stratified children with CSLD and compared their responses with 51 healthy control children. Cytokines involved in the generation and control of inflammation (IFN-γ, IL-13, IL-5, IL-10 at 72 hours and TNFα, IL-6, IL-10 at 24 hours) were measured in peripheral blood mononuclear cells challenged in vitro with live NTHi. We also measured circulating IgG subclass antibodies (IgG1 and IgG4) to two H. influenzae outer membrane proteins, P4 and P6. The most notable finding was that PBMC from children with CSLD produced significantly less IFN-γ in response to NTHi than healthy control children whereas mitogen-induced IFN-γ production was similar in both groups. Overall there were minor differences in innate and humoral immune responses between CSLD and control children. This study demonstrates that children with chronic suppurative lung disease have an altered systemic cell-mediated immune response to NTHi in vitro. This deficient IFN-γ response may contribute to increased susceptibility to NTHi infections and the pathogenesis of CSLD in children.
Collapse
Affiliation(s)
- Susan J. Pizzutto
- Menzies School of Health Research, Charles Darwin University, Brinkin, Northern Territory, Australia
- * E-mail:
| | - Stephanie T. Yerkovich
- Menzies School of Health Research, Charles Darwin University, Brinkin, Northern Territory, Australia
- Queensland Lung Transplant Service, The Prince Charles Hospital, Chermside, Queensland, Australia
- School of Medicine, The University of Queensland, St Lucia, Queensland, Australia
| | - John W. Upham
- School of Medicine, The University of Queensland, St Lucia, Queensland, Australia
- Department of Respiratory Medicine, Princess Alexandra Hospital, Wooloongabba, Queensland, Australia
| | - Belinda J. Hales
- Telethon Kids Institute, The University of Western Australia, Crawley, Western Australia, Australia
| | - Wayne R. Thomas
- Telethon Kids Institute, The University of Western Australia, Crawley, Western Australia, Australia
| | - Anne B. Chang
- Menzies School of Health Research, Charles Darwin University, Brinkin, Northern Territory, Australia
- The Department of Respiratory Medicine, Royal Children's Hospital, Herston, Queensland, Australia
- Queensland Children's Medical Research Institute, Royal Children's Hospital, Herston, Queensland, Australia
| |
Collapse
|
24
|
Lombardi C, Fiocchi A, Raffetti E, Donato F, Canonica GW, Passalacqua G. Cross-sectional comparison of the characteristics of respiratory allergy in immigrants and Italian children. Pediatr Allergy Immunol 2014; 25:473-80. [PMID: 25171740 DOI: 10.1111/pai.12250] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/03/2014] [Indexed: 01/28/2023]
Abstract
BACKGROUND Immigrants represent a good epidemiological model to evaluate the relative influence of environmental and inherited factors on the development of allergy. Several studies on allergy in adults have been published, but few data in children are available. We aimed to investigate the differences, between Italian and immigrant children, in clinical characteristics of respiratory allergy. METHODS This was a multicentre cross-sectional study involving children born in Italy from Italian parents and children born either in Italy or abroad from immigrants. Children referred firstly for allergic respiratory disease (rhinitis/asthma), with an ascertained clinical diagnosis and IgE sensitization to inhalants, were included. Demographic features, comorbidities, severity of disease, and sensitization profile were compared between Italians and immigrants, separating also those born in Italy from immigrant parents and those born abroad. RESULTS One hundred and sixty-five immigrant allergic children were enrolled (100 male, mean age 8.3 yr), 128 of whose had both parents immigrated. Italian children were 237 (156 male, mean age 8.4 yr). The Italian and immigrant children were similar, apart from pet's ownership and family size. There was no difference in the severity of rhinitis/asthma between the groups, whereas significant differences were found in the pattern of sensitization: immigrant children were more frequently sensitized to house dust mites (73.3% vs. 51%, respectively; p = 0.002) and less to grass (41.8% vs. 57.8%; p = 0.002); this was retained also in monosensitized children. Immigrant children born in Italy (n = 105) had a lower prevalence of rhinitis vs. Italians (68.3% vs. 87.6%, respectively, p = 0.003) and of sensitization to grass (28.3% vs. 49.5%, respectively, p = 0.008). No difference was found among macro-regions of origin and demographic or clinical features. CONCLUSIONS Immigrant children born either in Italy or abroad did not show significant differences in the clinical pattern of the respiratory allergic disease when compared to children born from Italian parents.
Collapse
Affiliation(s)
- Carlo Lombardi
- Pneumo-Allergologic Unit, Department of Internal Medicine and Geriatry, Poliambulanza Hospital, Brescia, Italy
| | | | | | | | | | | | | |
Collapse
|
25
|
Forsberg A, Abrahamsson TR, Björkstén B, Jenmalm MC. Pre- and postnatal administration of Lactobacillus reuteri decreases TLR2 responses in infants. Clin Transl Allergy 2014; 4:21. [PMID: 25002964 PMCID: PMC4083862 DOI: 10.1186/2045-7022-4-21] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 06/22/2014] [Indexed: 11/30/2022] Open
Abstract
Background Mice models indicate that intact Toll like receptor (TLR) signaling may be essential for the allergy protective effects of diverse bacterial exposure observed in clinical trials and epidemiological studies. Probiotic supplementation with Lactobacillus reuteri from pregnancy week 36 and to the infant through the first year of life decreased the prevalence of IgE-associated eczema at two years (ClinicalTrials.gov NCT01285830). The effect of this supplementation on innate immune responses to bacterial products and the expression of associated TLRs were explored. Methods Blood mononuclear cells were collected at birth, 6, 12 and 24 months from 61 infants and cultured with TLR2, 4 and 9 ligands. Cytokine and chemokine secretion was determined as well as TLR2, 4 and 9 mRNA expression. Results Probiotic supplementation was associated with decreased LTA (lipoteichoic acid) induced CCL4, CXCL8, IL-1β and IL-6 responses at 12 months and decreased CCL4 and IL-1β secretion at 24 months. TLR2 mRNA expression was not affected by probiotic treatment. Conclusions Decreased responses to TLR2, the main receptor for LTA from Gram positive bacteria, in probiotic treated children seem to be dependent on factors downstream of TLR mRNA expression.
Collapse
Affiliation(s)
- Anna Forsberg
- Department of Clinical and Experimental Medicine, Unit of Autoimmunity and Immune Regulation, Division of Clinical Immunology, Linköping University, SE-581 85 Linköping, Sweden
| | - Thomas R Abrahamsson
- Department of Clinical and Experimental Medicine, Division of Pediatrics, Linköping University, Linköping, Sweden
| | - Bengt Björkstén
- Institute of Environmental Medicine, Karolinska Institutet, Solna, Stockholm, Sweden ; School of Health and Medical Sciences, Örebro University, Örebro, Sweden
| | - Maria C Jenmalm
- Department of Clinical and Experimental Medicine, Unit of Autoimmunity and Immune Regulation, Division of Clinical Immunology, Linköping University, SE-581 85 Linköping, Sweden
| |
Collapse
|
26
|
Dowling DJ, Levy O. Ontogeny of early life immunity. Trends Immunol 2014; 35:299-310. [PMID: 24880460 DOI: 10.1016/j.it.2014.04.007] [Citation(s) in RCA: 268] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 04/29/2014] [Accepted: 04/30/2014] [Indexed: 12/18/2022]
Abstract
The human immune system comprises cellular and molecular components designed to coordinately prevent infection while avoiding potentially harmful inflammation and autoimmunity. Immunity varies with age, reflecting unique age-dependent challenges including fetal gestation, the neonatal phase, and infancy. Here, we review novel mechanistic insights into early life immunity, with an emphasis on emerging models of human immune ontogeny, which may inform age-specific translational development of novel anti-infectives, immunomodulators, and vaccines.
Collapse
Affiliation(s)
- David J Dowling
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Ofer Levy
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
27
|
Ng RLX, Scott NM, Bisley JL, Lambert MJ, Gorman S, Norval M, Hart PH. Characterization of regulatory dendritic cells differentiated from the bone marrow of UV-irradiated mice. Immunology 2014; 140:399-412. [PMID: 23826713 DOI: 10.1111/imm.12145] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 06/28/2013] [Accepted: 07/01/2013] [Indexed: 12/14/2022] Open
Abstract
When antigen-loaded dendritic cells (DCs) differentiated from the bone marrow (BM) of UV-irradiated mice (UV-BMDCs) were adoptively transferred into naive mice or mice pre-sensitized with that antigen, the recipients exhibited a reduced immune response following antigen challenge. Hence, UV-BMDCs are poorly immunogenic and can suppress pre-existing immunity. The UV-induced effect on BM-derived DCs was rapid (observed 1 day after UV radiation), long-lasting (observed 10 days after UV radiation) and UV dose-dependent. The mechanism by which UV-BMDCs could regulate immunity was investigated. The CD11c(+) cells, differentiated using granulocyte-macrophage colony-stimulating factor + interleukin-4, were confirmed to be DCs because they did not express the myeloid-derived suppressor cell marker, Gr1. UV-BMDCs did not display altered antigen uptake, processing or ability to activate T cells in vitro. When gene expression in UV-BMDCs and DCs differentiated from the BM of non-irradiated mice (control-BMDCs) was examined, Ccl7, Ccl8 and CSF1R (CD115) mRNA transcripts were up-regulated in UV-BMDCs compared with control-BMDCs. However, neutralizing antibodies for Ccl7 and Ccl8 did not abrogate the reduced immunogenicity of UV-BMDCs in vivo. Moreover, the up-regulation of CSF1R transcript did not correspond with increased receptor expression on UV-BMDCs. The phenotypes of UV-BMDCs and control-BMDCs were similar, with no difference in the expression of CD4, CD8α, CD103, B220 or F4/80, or the regulatory molecules CCR7 (CD197), FasL (CD95L), B7H3 (CD276) and B7H4. However, PDL1 (CD274) expression was reduced in UV-BMDCs compared with control-BMDCs following lipopolysaccharide stimulation. In summary, UV-BMDCs do not express the classical phenotypic or gene expression properties of DCs reported by others as 'regulatory' or 'tolerogenic'.
Collapse
Affiliation(s)
- Royce L X Ng
- Telethon Institute for Child Health Research, Centre for Child Health Research, University of Western Australia, Perth, WA, Australia
| | | | | | | | | | | | | |
Collapse
|
28
|
Smolen KK, Ruck CE, Fortuno ES, Ho K, Dimitriu P, Mohn WW, Speert DP, Cooper PJ, Esser M, Goetghebuer T, Marchant A, Kollmann TR. Pattern recognition receptor-mediated cytokine response in infants across 4 continents. J Allergy Clin Immunol 2013; 133:818-26.e4. [PMID: 24290283 PMCID: PMC3969582 DOI: 10.1016/j.jaci.2013.09.038] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 09/16/2013] [Accepted: 09/26/2013] [Indexed: 12/14/2022]
Abstract
Background Susceptibility to infection as well as response to vaccination varies among populations. To date, the underlying mechanisms responsible for these clinical observations have not been fully delineated. Because innate immunity instructs adaptive immunity, we hypothesized that differences between populations in innate immune responses may represent a mechanistic link to variation in susceptibility to infection or response to vaccination. Objective Determine whether differences in innate immune responses exist among infants from different continents of the world. Methods We determined the innate cytokine response following pattern recognition receptor (PRR) stimulation of whole blood from 2-year-old infants across 4 continents (Africa, North America, South America, and Europe). Results We found that despite the many possible genetic and environmental exposure differences in infants across 4 continents, innate cytokine responses were similar for infants from North America, South America, and Europe. However, cells from South African infants secreted significantly lower levels of cytokines than did cells from infants from the 3 other sites, and did so following stimulation of extracellular and endosomal but not cytosolic PRRs. Conclusions Substantial differences in innate cytokine responses to PRR stimulation exist among different populations of infants that could not have been predicted. Delineating the underlying mechanism(s) for these differences will not only aid in improving vaccine-mediated protection but possibly also provide clues for the susceptibility to infection in different regions of the world.
Collapse
Affiliation(s)
- Kinga K Smolen
- Department of Experimental Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Candice E Ruck
- Department of Experimental Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Edgardo S Fortuno
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kevin Ho
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Pedro Dimitriu
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - William W Mohn
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - David P Speert
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada; Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Philip J Cooper
- Centro de Investigaciones FEPIS, Esmeraldas, Quininde, Ecuador; Molecular and Biochemical Parasitology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, United Kingdom; Centro de Investgación en Enfermedades Infecciosas, Escuela de Biología, Pontificia Universidad Católica del Ecuador, Casilla, Quito, Ecuador
| | - Monika Esser
- Immunology Unit, Division of Medical Microbiology, Department of Pathology, NHLS and Stellenbosch University, Matieland, South Africa
| | - Tessa Goetghebuer
- Department of Paediatrics, Centre Hospitalier Universitaire Saint-Pierre, Brussels, Belgium
| | - Arnaud Marchant
- Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies, Belgium
| | - Tobias R Kollmann
- Department of Experimental Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
29
|
Microbial influence on tolerance and opportunities for intervention with prebiotics/probiotics and bacterial lysates. J Allergy Clin Immunol 2013; 131:1453-63; quiz 1464. [DOI: 10.1016/j.jaci.2013.03.020] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 02/19/2013] [Accepted: 03/22/2013] [Indexed: 01/21/2023]
|
30
|
Kollmann TR. Variation between Populations in the Innate Immune Response to Vaccine Adjuvants. Front Immunol 2013; 4:81. [PMID: 23565115 PMCID: PMC3613898 DOI: 10.3389/fimmu.2013.00081] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 03/18/2013] [Indexed: 12/15/2022] Open
Abstract
The success of the World Health Organization recommended “Expanded Program of Immunization” (EPI) and similar regional or national programs has been astounding. However, infectious threats currently not covered by these programs continue to infect millions of infants around the world. Furthermore, many infants do not receive existing vaccines either on time or for the required number of doses to provide optimal protection. Nor do all infants around the world develop the same protective immune response to the same vaccine. As a result approximately three million infants die every year from vaccine preventable infections. To tackle these issues, new vaccines need to be developed as well as existing ones made easier to administer. This requires identification of age-optimized vaccine schedules and formulations. In order to be most effective this approach will need to take population-based differences in response to vaccines and adjuvants into account. This review summarizes what is currently known about differences between populations around the world in the innate immune response to existing as well as new and promising vaccine adjuvants.
Collapse
Affiliation(s)
- Tobias R Kollmann
- Division of Infectious and Immunological Diseases, Department of Paediatrics, University of British Columbia Vancouver, BC, Canada
| |
Collapse
|
31
|
Pfefferle PI, von Mutius E. Hygiene and the cytokine jungle in Brazil. J Allergy Clin Immunol 2013; 131:1417-8. [PMID: 23545273 DOI: 10.1016/j.jaci.2013.02.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 02/27/2013] [Indexed: 10/27/2022]
|
32
|
Peden DB, Bush RK. Advances in environmental and occupational disorders in 2012. J Allergy Clin Immunol 2013; 131:668-74. [PMID: 23384680 DOI: 10.1016/j.jaci.2012.12.1572] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Accepted: 12/27/2012] [Indexed: 10/27/2022]
Abstract
The year 2012 produced a number of advances in our understanding of the effect of environmental factors on allergic diseases, identification of new allergens, immune mechanisms in host defense, factors involved in asthma severity, and therapeutic approaches. This review focuses on the articles published in the Journal in 2012 that enhance our knowledge base of environmental and occupational disorders. Identification of novel allergens can improve diagnostics, risk factor analysis can aid preventative approaches, and studies of genetic-environmental interactions and immune mechanisms will lead to better therapeutics.
Collapse
Affiliation(s)
- David B Peden
- Department of Pediatrics, Division of Allergy, Immunology, Rheumatology, and Infectious Diseases, School of Medicine, University of North Carolina, Chapel Hill, NC 27599-7310, USA.
| | | |
Collapse
|