1
|
Hillson K, Saglani S, Custovic A. Preschool wheeze and asthma endotypes- implications for future therapy. Expert Rev Respir Med 2024; 18:1025-1039. [PMID: 39655566 DOI: 10.1080/17476348.2024.2440468] [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: 05/12/2024] [Accepted: 12/06/2024] [Indexed: 12/14/2024]
Abstract
INTRODUCTION Preschool wheeze and school-aged asthma present a large healthcare burden. Both conditions are now recognized to be heterogeneous, with similar symptom presentation but likely different underlying lung pathology. AREAS COVERED Current treatment options for preschool wheeze are constrained by extrapolations from the management of school-aged children with asthma. While most cases of asthma at school age are caused by classical atopic, eosinophilic, Type-2 driven asthma, only a quarter of preschool children with wheeze fall into this category. Targeting treatment to specific underlying mechanisms resulting in preschool wheeze may alter the progression to school age asthma. Novel biologics have revolutionized the management of severe, treatment-resistant school age asthma, but a limited evidence base limits their use in young children. There are several potential future non-steroid-based treatment options in development, of which bacterial lysates show the most promise. EXPERT OPINION Effective treatment of preschool wheeze may preserve lung function into later life, which may alter the progression trajectory toward school age asthma. Endotype-driven management will enable more effective treatment of both preschool wheeze and school age asthma.
Collapse
Affiliation(s)
- Kushalinii Hillson
- National Heart and Lung Institute, Imperial College London, London, UK
- Paediatric Respiratory Medicine Department, Royal Brompton Hospital, London, UK
| | - Sejal Saglani
- National Heart and Lung Institute, Imperial College London, London, UK
- Paediatric Respiratory Medicine Department, Royal Brompton Hospital, London, UK
- NIHR Imperial Biomedical Research Centre (BRC), London, UK
| | - Adnan Custovic
- National Heart and Lung Institute, Imperial College London, London, UK
- NIHR Imperial Biomedical Research Centre (BRC), London, UK
| |
Collapse
|
2
|
Rahman MM, Grice ID, Ulett GC, Wei MQ. Advances in Bacterial Lysate Immunotherapy for Infectious Diseases and Cancer. J Immunol Res 2024; 2024:4312908. [PMID: 38962577 PMCID: PMC11221958 DOI: 10.1155/2024/4312908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/22/2024] [Accepted: 04/26/2024] [Indexed: 07/05/2024] Open
Abstract
Antigenic cell fragments, pathogen-associated molecular patterns, and other immunostimulants in bacterial lysates or extracts may induce local and systemic immune responses in specific and nonspecific paradigms. Based on current knowledge, this review aimed to determine whether bacterial lysate has comparable functions in infectious diseases and cancer treatment. In infectious diseases, including respiratory and urinary tract infections, immune system activation by bacterial lysate can identify and combat pathogens. Commercially available bacterial lysates, including OM-85, Ismigen, Lantigen B, and LW 50020, were effective in children and adults in treating respiratory tract infections, chronic obstructive pulmonary disease, rhinitis, and rhinosinusitis with varying degrees of success. Moreover, OM-89, Uromune, Urovac, Urivac, and ExPEC4V showed therapeutic benefits in controlling urinary tract infections in adults, especially women. Bacterial lysate-based therapeutics are safe, well-tolerated, and have few side effects, making them a good alternative for infectious disease management. Furthermore, a nonspecific immunomodulation by bacterial lysates may stimulate innate immunity, benefiting cancer treatment. "Coley's vaccine" has been used to treat sarcomas, carcinomas, lymphomas, melanomas, and myelomas with varying outcomes. Later, several similar bacterial lysate-based therapeutics have been developed to treat cancers, including bladder cancer, non-small cell lung cancer, and myeloma; among them, BCG for in situ bladder cancer is well-known. Proinflammatory cytokines, including IL-1, IL-6, IL-12, and TNF-α, may activate bacterial antigen-specific adaptive responses that could restore tumor antigen recognition and response by tumor-specific type 1 helper cells and cytotoxic T cells; therefore, bacterial lysates are worth investigating as a vaccination adjuvants or add-on therapies for several cancers.
Collapse
Affiliation(s)
- Md. Mijanur Rahman
- School of Pharmacy and Medical SciencesGriffith University, Gold Coast 4222, QLD, Australia
- Menzies Health Institute QueenslandGriffith University, Gold Coast 4222, QLD, Australia
| | - I. Darren Grice
- School of Pharmacy and Medical SciencesGriffith University, Gold Coast 4222, QLD, Australia
- Institute for GlycomicsGriffith University, Gold Coast 4222, QLD, Australia
| | - Glen C. Ulett
- School of Pharmacy and Medical SciencesGriffith University, Gold Coast 4222, QLD, Australia
- Menzies Health Institute QueenslandGriffith University, Gold Coast 4222, QLD, Australia
| | - Ming Q. Wei
- School of Pharmacy and Medical SciencesGriffith University, Gold Coast 4222, QLD, Australia
- Menzies Health Institute QueenslandGriffith University, Gold Coast 4222, QLD, Australia
| |
Collapse
|
3
|
Grijincu M, Buzan MR, Zbîrcea LE, Păunescu V, Panaitescu C. Prenatal Factors in the Development of Allergic Diseases. Int J Mol Sci 2024; 25:6359. [PMID: 38928067 PMCID: PMC11204337 DOI: 10.3390/ijms25126359] [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: 05/06/2024] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Allergic diseases are showing increasing prevalence in Western societies. They are characterized by a heightened reactivity towards otherwise harmless environmental stimuli. Allergic diseases showing a wide range of severity of symptoms have a significant impact on the quality of life of affected individuals. This study aims to highlight the mechanisms that induce these reactions, how they progress, and which prenatal factors influence their development. Most frequently, the reaction is mediated by immunoglobulin E (IgE) produced by B cells, which binds to the surface of mast cells and basophils and triggers an inflammatory response. The antibody response is triggered by a shift in T-cell immune response. The symptoms often start in early childhood with eczema or atopic dermatitis and progress to allergic asthma in adolescence. An important determinant of allergic diseases seems to be parental, especially maternal history of allergy. Around 30% of children of allergic mothers develop allergic sensitization in childhood. Genes involved in the regulation of the epithelial barrier function and the T-cell response were found to affect the predisposition to developing allergic disorders. Cord blood IgE was found to be a promising predictor of allergic disease development. Fetal B cells produce IgE starting at the 20th gestation week. These fetal B cells could be sensitized together with mast cells by maternal IgE and IgE-allergen complexes crossing the placental barrier via the low-affinity IgE receptor. Various factors were found to facilitate these sensitizations, including pesticides, drugs, exposure to cigarette smoke and maternal uncontrolled asthma. Prenatal exposure to microbial infections and maternal IgG appeared to play a role in the regulation of T-cell response, indicating a protective effect against allergy development. Additional preventive factors were dietary intake of vitamin D and omega 3 fatty acids as well as decreased maternal IgE levels. The effect of exposure to food allergens during pregnancy was inconclusive, with studies having found both sensitizing and protective effects. In conclusion, prenatal factors including genetics, epigenetics and fetal environmental factors have an important role in the development of allergic disorders in later life. Children with a genetic predisposition are at risk when exposed to cigarette smoke as well as increased maternal IgE in the prenatal period. Maternal diet during pregnancy and immunization against certain allergens could help in the prevention of allergy in predisposed children.
Collapse
Affiliation(s)
- Manuela Grijincu
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, Victor Babeș University of Medicine and Pharmacy, 300041 Timișoara, Romania
- OncoGen Center, Pius Brînzeu County Clinical Emergency Hospital, 300723 Timișoara, Romania
| | - Maria-Roxana Buzan
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, Victor Babeș University of Medicine and Pharmacy, 300041 Timișoara, Romania
- OncoGen Center, Pius Brînzeu County Clinical Emergency Hospital, 300723 Timișoara, Romania
| | - Lauriana-Eunice Zbîrcea
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, Victor Babeș University of Medicine and Pharmacy, 300041 Timișoara, Romania
- OncoGen Center, Pius Brînzeu County Clinical Emergency Hospital, 300723 Timișoara, Romania
| | - Virgil Păunescu
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, Victor Babeș University of Medicine and Pharmacy, 300041 Timișoara, Romania
- OncoGen Center, Pius Brînzeu County Clinical Emergency Hospital, 300723 Timișoara, Romania
| | - Carmen Panaitescu
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, Victor Babeș University of Medicine and Pharmacy, 300041 Timișoara, Romania
- OncoGen Center, Pius Brînzeu County Clinical Emergency Hospital, 300723 Timișoara, Romania
| |
Collapse
|
4
|
Rahbek-Hansen SH, Mikkelsen M, Stokholm J, Bønnelykke K, Chawes BL, Brustad N. Preventive effects of prenatal administration of OM-85/BV on asthma and respiratory infection risk in the offspring: A review of animal models. Pediatr Allergy Immunol 2024; 35:e14184. [PMID: 38924159 DOI: 10.1111/pai.14184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024]
Abstract
Asthma is the most common chronic disease in childhood affecting the daily lives of many patients despite current treatment regimens. Therefore, the need for new therapeutic approaches is evident, where a primary prevention strategy is the ultimate goal. Studies of children born to mothers in farming environments have shown a lower risk of respiratory infections and asthma development. Already at birth, these newborns have demonstrated accelerated maturation and upregulation of host defense immune functions suggesting a prenatal transplacental training of the innate immune system through maternal microbial exposure. This mechanism could possibly be utilized to help prevent both respiratory infections and asthma in young children. Human studies exploring the potential preventative effects of pregnancy bacterial lysate treatment on asthma and respiratory infections are lacking, however, this has been studied in experimental studies using mice through administrations of the bacterial lysate OM-85. This review will present the current literature on the immunomodulatory effects relevant for respiratory infections and asthma in the offspring of mice treated with OM-85 throughout pregnancy. Further, the review will discuss the cellular and molecular mechanisms behind these effects. In conclusion, we found promising results of an accelerated immune competence and improved resistance to airway challenges as a result of prenatal bacterial lysate treatment that may pave the way for implementing this in human trials to prevent asthma and respiratory infections.
Collapse
Affiliation(s)
- Signe Hahn Rahbek-Hansen
- Copenhagen Prospective Studies on Asthma in Childhood, Health Sciences, Danish Pediatric Asthma Center, Copenhagen University Hospital, University of Copenhagen, Gentofte, Denmark
| | - Marianne Mikkelsen
- Copenhagen Prospective Studies on Asthma in Childhood, Health Sciences, Danish Pediatric Asthma Center, Copenhagen University Hospital, University of Copenhagen, Gentofte, Denmark
| | - Jakob Stokholm
- Copenhagen Prospective Studies on Asthma in Childhood, Health Sciences, Danish Pediatric Asthma Center, Copenhagen University Hospital, University of Copenhagen, Gentofte, Denmark
| | - Klaus Bønnelykke
- Copenhagen Prospective Studies on Asthma in Childhood, Health Sciences, Danish Pediatric Asthma Center, Copenhagen University Hospital, University of Copenhagen, Gentofte, Denmark
| | - Bo L Chawes
- Copenhagen Prospective Studies on Asthma in Childhood, Health Sciences, Danish Pediatric Asthma Center, Copenhagen University Hospital, University of Copenhagen, Gentofte, Denmark
| | - Nicklas Brustad
- Copenhagen Prospective Studies on Asthma in Childhood, Health Sciences, Danish Pediatric Asthma Center, Copenhagen University Hospital, University of Copenhagen, Gentofte, Denmark
| |
Collapse
|
5
|
Abstract
Allergic diseases typically begin in early life and can impose a heavy burden on children and their families. Effective preventive measures are currently unavailable but may be ushered in by studies on the "farm effect", the strong protection from asthma and allergy found in children born and raised on traditional farms. Two decades of epidemiologic and immunologic research have demonstrated that this protection is provided by early and intense exposure to farm-associated microbes that target primarily innate immune pathways. Farm exposure also promotes timely maturation of the gut microbiome, which mediates a proportion of the protection conferred by the farm effect. Current research seeks to identify allergy-protective compounds from traditional farm environments, but standardization and regulation of such substances will likely prove challenging. On the other hand, studies in mouse models show that administration of standardized, pharmacological-grade lysates of human airway bacteria abrogates allergic lung inflammation by acting on multiple innate immune targets, including the airway epithelium/IL-33/ILC2 axis and dendritic cells whose Myd88/Trif-dependent tolerogenic reprogramming is sufficient for asthma protection in adoptive transfer models. To the extent that these bacterial lysates mimic the protective effects of natural exposure to microbe-rich environments, these agents might provide an effective tool for prevention of allergic disease.
Collapse
Affiliation(s)
- Donata Vercelli
- Department of Cellular and Molecular Medicine, Asthma & Airway Disease Research Center, The BIO5 Institute, and The Arizona Center for the Biology of Complex Diseases, The University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
6
|
Sikder MAA, Rashid RB, Ahmed T, Sebina I, Howard DR, Ullah MA, Rahman MM, Lynch JP, Curren B, Werder RB, Simpson J, Bissell A, Morrison M, Walpole C, Radford KJ, Kumar V, Woodruff TM, Ying TH, Ali A, Kaiko GE, Upham JW, Hoelzle RD, Cuív PÓ, Holt PG, Dennis PG, Phipps S. Maternal diet modulates the infant microbiome and intestinal Flt3L necessary for dendritic cell development and immunity to respiratory infection. Immunity 2023; 56:1098-1114.e10. [PMID: 37003256 DOI: 10.1016/j.immuni.2023.03.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 12/28/2022] [Accepted: 03/02/2023] [Indexed: 04/03/2023]
Abstract
Poor maternal diet during pregnancy is a risk factor for severe lower respiratory infections (sLRIs) in the offspring, but the underlying mechanisms remain elusive. Here, we demonstrate that in mice a maternal low-fiber diet (LFD) led to enhanced LRI severity in infants because of delayed plasmacytoid dendritic cell (pDC) recruitment and perturbation of regulatory T cell expansion in the lungs. LFD altered the composition of the maternal milk microbiome and assembling infant gut microbiome. These microbial changes reduced the secretion of the DC growth factor Flt3L by neonatal intestinal epithelial cells and impaired downstream pDC hematopoiesis. Therapy with a propionate-producing bacteria isolated from the milk of high-fiber diet-fed mothers, or supplementation with propionate, conferred protection against sLRI by restoring gut Flt3L expression and pDC hematopoiesis. Our findings identify a microbiome-dependent Flt3L axis in the gut that promotes pDC hematopoiesis in early life and confers disease resistance against sLRIs.
Collapse
Affiliation(s)
- Md Al Amin Sikder
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh
| | - Ridwan B Rashid
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Tufael Ahmed
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia; School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Ismail Sebina
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia
| | - Daniel R Howard
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Md Ashik Ullah
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia
| | - Muhammed Mahfuzur Rahman
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jason P Lynch
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia
| | - Bodie Curren
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia
| | - Rhiannon B Werder
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia
| | - Jennifer Simpson
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia; School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle, Newcastle, NSW, Australia
| | - Alec Bissell
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia
| | - Mark Morrison
- University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, Brisbane, QLD 4102, Australia; Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Carina Walpole
- Mater Research Institute, The University of Queensland, Translational Research Institute, Wolloongabba, Brisbane, QLD 4102, Australia
| | - Kristen J Radford
- Mater Research Institute, The University of Queensland, Translational Research Institute, Wolloongabba, Brisbane, QLD 4102, Australia
| | - Vinod Kumar
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia; Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Tan Hui Ying
- School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Ayesha Ali
- School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Gerard E Kaiko
- School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - John W Upham
- University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, Brisbane, QLD 4102, Australia; Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia; Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Robert D Hoelzle
- The School of Earth and Environmental Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Páraic Ó Cuív
- Mater Research Institute, The University of Queensland, Translational Research Institute, Wolloongabba, Brisbane, QLD 4102, Australia; Microba Life Sciences, Translational Research Institute, Woolloongabba, Brisbane, QLD 4102, Australia
| | - Patrick G Holt
- Telethon Kids Institute, The University of Western Australia, Nedlands, WA 6009, Australia
| | - Paul G Dennis
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia; The School of Earth and Environmental Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Simon Phipps
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia; School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD 4000, Australia; Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
7
|
Pivniouk V, Vercelli D. The OM-85 bacterial lysate: a new tool against SARS-CoV-2? Multidiscip Respir Med 2023; 18:906. [PMID: 36798954 PMCID: PMC9926922 DOI: 10.4081/mrm.2023.906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 01/05/2023] [Indexed: 01/27/2023] Open
Abstract
The emergence of SARS-CoV-2, a novel coronavirus, caused the global Coronavirus disease of 2019 (COVID-19) pandemic. Because SARS-CoV-2 mutates rapidly, vaccines that induce immune responses against viral components critical for target cell infection strongly mitigate but do not abrogate viral spread, and disease rates remain high worldwide. Complementary treatments are therefore needed to reduce the frequency and/or severity of SARS-CoV-2 infections. OM-85, a standardized lysate of 21 bacterial strains often found in the human airways, has immuno-modulatory properties and is widely used empirically in Europe, South America and Asia for the prophylaxis of recurrent upper airway infections in adults and children, with excellent safety profiles. In vitro studies from our laboratory recently demonstrated that OM-85 inhibits SARS-CoV-2 epithelial cell infection by downregulating SARS-CoV-2 receptor expression, raising the possibility that this bacterial extract might eventually complement the current COVID-19 therapeutic toolkit. Here we discuss how our results and those from other groups are fostering progress in this emerging field of research.
Collapse
Affiliation(s)
- Vadim Pivniouk
- Department of Cellular and Molecular Medicine
- Asthma and Airway Disease Research Center
| | - Donata Vercelli
- Department of Cellular and Molecular Medicine
- Asthma and Airway Disease Research Center
- The BIO5 Institute
- Arizona Center for the Biology of Complex Diseases, The University of Arizona, Tucson, AZ, USA
| |
Collapse
|
8
|
DeVries A, McCauley K, Fadrosh D, Fujimura KE, Stern DA, Lynch SV, Vercelli D. Maternal prenatal immunity, neonatal trained immunity, and early airway microbiota shape childhood asthma development. Allergy 2022; 77:3617-3628. [PMID: 35841380 PMCID: PMC9712226 DOI: 10.1111/all.15442] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 05/25/2022] [Accepted: 06/11/2022] [Indexed: 01/28/2023]
Abstract
BACKGROUND The path to childhood asthma is thought to initiate in utero and be further promoted by postnatal exposures. However, the underlying mechanisms remain underexplored. We hypothesized that prenatal maternal immune dysfunction associated with increased childhood asthma risk (revealed by low IFN-γ:IL-13 secretion during the third trimester of pregnancy) alters neonatal immune training through epigenetic mechanisms and promotes early-life airway colonization by asthmagenic microbiota. METHODS We examined epigenetic, immunologic, and microbial features potentially related to maternal prenatal immunity (IFN-γ:IL-13 ratio) and childhood asthma in a birth cohort of mother-child dyads sampled pre-, peri-, and postnatally (N = 155). Epigenome-wide DNA methylation and cytokine production were assessed in cord blood mononuclear cells (CBMC) by array profiling and ELISA, respectively. Nasopharyngeal microbiome composition was characterized at age 2-36 months by 16S rRNA sequencing. RESULTS Maternal prenatal immune status related to methylome profiles in neonates born to non-asthmatic mothers. A module of differentially methylated CpG sites enriched for microbe-responsive elements was associated with childhood asthma. In vitro responsiveness to microbial products was impaired in CBMCs from neonates born to mothers with the lowest IFN-γ:IL-13 ratio, suggesting defective neonatal innate immunity in those who developed asthma during childhood. These infants exhibited a distinct pattern of upper airway microbiota development characterized by early-life colonization by Haemophilus that transitioned to a Moraxella-dominated microbiota by age 36 months. CONCLUSIONS Maternal prenatal immune status shapes asthma development in her child by altering the epigenome and trained innate immunity at birth, and is associated with pathologic upper airway microbial colonization in early life.
Collapse
Affiliation(s)
- Avery DeVries
- Asthma and Airway Disease Research CenterThe University of ArizonaTucsonArizonaUSA
- The BIO5 InstituteThe University of ArizonaTucsonArizonaUSA
| | - Kathryn McCauley
- Department of MedicineUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Benioff Center for Microbiome MedicineUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Douglas Fadrosh
- Department of MedicineUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Kei E. Fujimura
- Genetic Disease LabCalifornia Department of Public HealthRichmondCaliforniaUSA
| | - Debra A. Stern
- Asthma and Airway Disease Research CenterThe University of ArizonaTucsonArizonaUSA
| | - Susan V. Lynch
- Department of MedicineUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Benioff Center for Microbiome MedicineUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Donata Vercelli
- Asthma and Airway Disease Research CenterThe University of ArizonaTucsonArizonaUSA
- The BIO5 InstituteThe University of ArizonaTucsonArizonaUSA
- Department of Cellular and Molecular MedicineThe University of ArizonaTucsonArizonaUSA
- Arizona Center for the Biology of Complex DiseasesThe University of ArizonaTucsonArizonaUSA
| |
Collapse
|
9
|
Fortmann MI, Dirks J, Goedicke-Fritz S, Liese J, Zemlin M, Morbach H, Härtel C. Immunization of preterm infants: current evidence and future strategies to individualized approaches. Semin Immunopathol 2022; 44:767-784. [PMID: 35922638 PMCID: PMC9362650 DOI: 10.1007/s00281-022-00957-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 07/08/2022] [Indexed: 12/15/2022]
Abstract
Preterm infants are at particularly high risk for infectious diseases. As this vulnerability extends beyond the neonatal period into childhood and adolescence, preterm infants benefit greatly from infection-preventive measures such as immunizations. However, there is an ongoing discussion about vaccine safety and efficacy due to preterm infants' distinct immunological features. A significant proportion of infants remains un- or under-immunized when discharged from primary hospital stay. Educating health care professionals and parents, promoting maternal immunization and evaluating the potential of new vaccination tools are important means to reduce the overall burden from infectious diseases in preterm infants. In this narrative review, we summarize the current knowledge about vaccinations in premature infants. We discuss the specificities of early life immunity and memory function, including the role of polyreactive B cells, restricted B cell receptor diversity and heterologous immunity mediated by a cross-reactive T cell repertoire. Recently, mechanistic studies indicated that tissue-resident memory (Trm) cell populations including T cells, B cells and macrophages are already established in the fetus. Their role in human early life immunity, however, is not yet understood. Tissue-resident memory T cells, for example, are diminished in airway tissues in neonates as compared to older children or adults. Hence, the ability to make specific recall responses after secondary infectious stimulus is hampered, a phenomenon that is transcriptionally regulated by enhanced expression of T-bet. Furthermore, the microbiome establishment is a dominant factor to shape resident immunity at mucosal surfaces, but it is often disturbed in the context of preterm birth. The proposed function of Trm T cells to remember benign interactions with the microbiome might therefore be reduced which would contribute to an increased risk for sustained inflammation. An improved understanding of Trm interactions may determine novel targets of vaccination, e.g., modulation of T-bet responses and facilitate more individualized approaches to protect preterm babies in the future.
Collapse
Affiliation(s)
- Mats Ingmar Fortmann
- Department of Pediatrics, University Lübeck, University Hospital Schleswig-Holstein Campus Lübeck, Lübeck, Germany
| | - Johannes Dirks
- Department of Pediatrics, University Hospital of Würzburg, Würzburg, Germany
| | - Sybelle Goedicke-Fritz
- Department of General Pediatrics and Neonatology, Faculty of Medicine, Saarland University Hospital and Saarland University, Homburg, Germany
| | - Johannes Liese
- Department of Pediatrics, University Hospital of Würzburg, Würzburg, Germany
| | - Michael Zemlin
- Department of General Pediatrics and Neonatology, Faculty of Medicine, Saarland University Hospital and Saarland University, Homburg, Germany
| | - Henner Morbach
- Department of General Pediatrics and Neonatology, Faculty of Medicine, Saarland University Hospital and Saarland University, Homburg, Germany
| | - Christoph Härtel
- Department of Pediatrics, University Hospital of Würzburg, Würzburg, Germany.
| |
Collapse
|
10
|
Ponsonby AL, Collier F, O'Hely M, Tang MLK, Ranganathan S, Gray L, Morwitch E, Saffery R, Burgner D, Dwyer T, Sly PD, Harrison LC, Vuillermin P. Household size, T regulatory cell development, and early allergic disease: a birth cohort study. Pediatr Allergy Immunol 2022; 33:e13810. [PMID: 35754137 PMCID: PMC9545943 DOI: 10.1111/pai.13810] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 05/05/2022] [Accepted: 05/19/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Children born to larger households have less allergic disease. T regulatory cell (Treg) development may be a relevant mechanism, but this has not been studied longitudinally. OBJECTIVE We aim to (i) describe how prenatal and postnatal environmental factors are associated with Treg development and (ii) investigate whether serial Treg measures predict allergic outcomes at 1 year of age. METHODS A birth cohort (n = 1074) with information on prenatal and postnatal early life factors. Both naïve Treg (nTreg) and activated Treg (aTreg) cell populations (as a proportion of CD4+ T cells) were available in 463 infants at birth (cord blood), 600 at 6 months, and 675 at 12 months. 191 infants had serial measures. Measures of allergic status at 12 months were polysensitization (sensitization to 2 or more allergens), clinically proven food allergy, atopic eczema, and atopic wheeze. RESULTS Infants born to larger households (3 or more residents) had higher longitudinal nTreg proportions over the first postnatal year with a mean difference (MD) of 0.67 (95% CI 0.30-1.04)%. Higher nTreg proportions at birth were associated with a reduced risk of infant allergic outcomes. Childcare attendance and breastfeeding were associated with higher longitudinal nTreg proportions (MD 0.48 (95% CI 0.08-0.80)%. CONCLUSION Multiple prenatal and postnatal microbial factors are associated with nTreg and aTreg development. Larger household size was associated with higher nTreg at birth which in turn was associated with reduced allergic sensitization and disease at 12 months of age.
Collapse
Affiliation(s)
- Anne-Louise Ponsonby
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia.,Murdoch Children's Research Institute, Royal Children's Hospital, University of Melbourne, Parkville, Victoria, Australia
| | - Fiona Collier
- School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Martin O'Hely
- Murdoch Children's Research Institute, Royal Children's Hospital, University of Melbourne, Parkville, Victoria, Australia.,School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Mimi L K Tang
- Murdoch Children's Research Institute, Royal Children's Hospital, University of Melbourne, Parkville, Victoria, Australia
| | - Sarath Ranganathan
- Murdoch Children's Research Institute, Royal Children's Hospital, University of Melbourne, Parkville, Victoria, Australia
| | - Lawrence Gray
- School of Medicine, Deakin University, Geelong, Victoria, Australia.,Barwon Health, Geelong, Victoria, Australia
| | - Ellen Morwitch
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia.,National Centre of Epidemiology and Population Health, Australian National University, Canberra, Australia
| | - Richard Saffery
- Murdoch Children's Research Institute, Royal Children's Hospital, University of Melbourne, Parkville, Victoria, Australia
| | - David Burgner
- Murdoch Children's Research Institute, Royal Children's Hospital, University of Melbourne, Parkville, Victoria, Australia
| | - Terence Dwyer
- Murdoch Children's Research Institute, Royal Children's Hospital, University of Melbourne, Parkville, Victoria, Australia.,The George Institute for Global Health, Oxford University, Oxford, UK
| | - Peter D Sly
- Murdoch Children's Research Institute, Royal Children's Hospital, University of Melbourne, Parkville, Victoria, Australia.,University of Queensland, South Brisbane, Queensland, Australia
| | | | - Peter Vuillermin
- Murdoch Children's Research Institute, Royal Children's Hospital, University of Melbourne, Parkville, Victoria, Australia.,School of Medicine, Deakin University, Geelong, Victoria, Australia.,Barwon Health, Geelong, Victoria, Australia
| | | |
Collapse
|
11
|
The extended farm effect: The milk protein β-lactoglobulin in stable dust protects against allergies. Allergol Select 2022; 6:111-117. [PMID: 35392214 PMCID: PMC8982089 DOI: 10.5414/alx02246e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 02/08/2022] [Indexed: 12/16/2022] Open
Abstract
Background: The allergy- and asthma-protective farm effect is mediated by numerous factors. Especially dust from cattle stables and raw cow’s milk show beneficial properties, suggesting a bovine protein to be involved. As a major milk protein and member of the lipocalin family, β-lactoglobulin (BLG) binds small, hydrophobic ligands and thereby modulates the immune response. Empty BLG promotes allergy development, whereas BLG in association with ligands shows allergy-preventive as well as allergy-reducing effects in vivo and in vitro. Results: BLG has been identified as a major protein in stable dust (therein bound to zinc) as well as in the air around cattle stables. This association with zinc favors an allergy-protective immune profile. Conclusion: Its immune-modulating, allergy-protective characteristics together with its presence in raw cow’s milk as well as in stable dust and ambient air render BLG an essential contributor to the farm effect.
Collapse
|
12
|
Rosenfeld N, Mandelberg A, Dalal I, Tasher D, Kamar A, Schnapper M, Armoni Domany K. The impact of the COVID-19 pandemic on respiratory morbidity during infancy: A birth-cohort study. Pediatr Pulmonol 2022; 57:848-856. [PMID: 35018744 DOI: 10.1002/ppul.25822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/04/2022] [Accepted: 01/08/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE To evaluate the incidence of wheezing and overall respiratory morbidity in healthy infants born during the first peak of the coronavirus disease-2019 (COVID-19) pandemic, compared with infants born during the preceding year. METHODS This was a single-center retrospective birth cohort study to compare a cohort of children born between February and March 2020 (COVID-19 group) to a control group of children born between February and March 2019 (pre-COVID-19 group). At 1 year of age, we collected respiratory data using parental and telephone questionnaires. PRIMARY OUTCOME wheezing incidence and/or bronchodilator use. SECONDARY OUTCOMES recurrent wheezing, emergency-room visits, hospital admissions, pneumonia diagnosis, and admissions due to lower-respiratory-tract-infections (LRTI). We included the following covariate risk factors in the logistic regression models; atopy, daycare attendance, breastmilk feeding, parental smoking, C-section, siblings, and gestational age. RESULTS We enrolled 588 infants, 294 in each group (48% males). Demographic, perinatal, and atopic characteristics were similar between the groups. Compared to the pre-COVID-19 group, infants born during the COVID-19 period were significantly less likely to report wheezing and/or bronchodilator use (adjusted-odds ratio [OR], 0.4; 95% confidence interval [CI] 0.28-0.59), systemic steroid use, (adjusted-OR, 0.47; 95% CI 0.24-0.91), emergency-room visits (adjusted-OR, 0.36; 95% CI 0.17-0.72), LRTI admissions (adjusted-OR, 0.2; 95% CI 0.05-0.74), or pneumonia diagnosis (adjusted-OR, 0.22; 95% CI 0.09-0.53). CONCLUSIONS This study investigated wheezing and respiratory morbidity over the first year of the COVID-19 pandemic in infants born during the first peak of COVID-19. The study demonstrated a significant decrease in most aspects of respiratory morbidity. A longitudinal follow-up study to explore the subsequent impact of these findings is warranted.
Collapse
Affiliation(s)
- Nataly Rosenfeld
- Department of Pediatric, The Edith Wolfson Medical Center, Holon, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Avigdor Mandelberg
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,Pediatric Pulmonology Unit, The Edith Wolfson Medical Center, Holon, Israel
| | - Ilan Dalal
- Department of Pediatric, The Edith Wolfson Medical Center, Holon, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Diana Tasher
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,Infectious Diseases Unit, The Edith Wolfson Medical Center, Holon, Israel
| | - Alma Kamar
- Department of Pediatric, The Edith Wolfson Medical Center, Holon, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Michael Schnapper
- Department of Pediatric, The Edith Wolfson Medical Center, Holon, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Keren Armoni Domany
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,Pediatric Pulmonology Unit, The Edith Wolfson Medical Center, Holon, Israel
| |
Collapse
|
13
|
Pivniouk V, Gimenes-Junior JA, Ezeh P, Michael A, Pivniouk O, Hahn S, VanLinden SR, Malone SP, Abidov A, Anderson D, Gozdz J, DeVries A, Martinez FD, Pasquali C, Vercelli D. Airway administration of OM-85, a bacterial lysate, blocks experimental asthma by targeting dendritic cells and the epithelium/IL-33/ILC2 axis. J Allergy Clin Immunol 2022; 149:943-956. [PMID: 34560105 PMCID: PMC8901455 DOI: 10.1016/j.jaci.2021.09.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 08/26/2021] [Accepted: 09/02/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Microbial interventions against allergic asthma have robust epidemiologic underpinnings and the potential to recalibrate disease-inducing immune responses. Oral administration of OM-85, a standardized lysate of human airways bacteria, is widely used empirically to prevent respiratory infections and a clinical trial is testing its ability to prevent asthma in high-risk children. We previously showed that intranasal administration of microbial products from farm environments abrogates experimental allergic asthma. OBJECTIVES We sought to investigate whether direct administration of OM-85 to the airway compartment protects against experimental allergic asthma; and to identify protective cellular and molecular mechanisms activated through this natural route. METHODS Different strains of mice sensitized and challenged with ovalbumin or Alternaria received OM-85 intranasally, and cardinal cellular and molecular asthma phenotypes were measured. Airway transfer experiments assessed whether OM-85-treated dendritic cells protect allergen-sensitized, OM-85-naive mice against asthma. RESULTS Airway OM-85 administration suppressed allergic asthma in all models acting on multiple innate and adaptive immune targets: the airway epithelium/IL-33/ILC2 axis, lung allergen-induced type 2 responses, and dendritic cells whose Myd88/Trif-dependent tolerogenic reprogramming was sufficient to transfer OM-85-induced asthma protection. CONCLUSIONS We provide the first demonstration that administering a standardized bacterial lysate to the airway compartment protects from experimental allergic asthma by engaging multiple immune pathways. Because protection required a cumulative dose 27- to 46-fold lower than the one reportedly active through the oral route, the efficacy of intranasal OM-85 administration may reflect its direct access to the airway mucosal networks controlling the initiation and development of allergic asthma.
Collapse
Affiliation(s)
- Vadim Pivniouk
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Ariz; Asthma and Airway Disease Research Center, The University of Arizona, Tucson, Ariz; BIO5 Institute, The University of Arizona, Tucson, Ariz.
| | - Joao A. Gimenes-Junior
- Asthma and Airway Disease Research Center, The University
of Arizona, Tucson, AZ, 85721, USA
| | - Peace Ezeh
- Asthma and Airway Disease Research Center, The University
of Arizona, Tucson, AZ, 85721, USA
| | - Ashley Michael
- Asthma and Airway Disease Research Center, The University
of Arizona, Tucson, AZ, 85721, USA
| | - Oksana Pivniouk
- Asthma and Airway Disease Research Center, The University
of Arizona, Tucson, AZ, 85721, USA
| | - Seongmin Hahn
- Asthma and Airway Disease Research Center, The University
of Arizona, Tucson, AZ, 85721, USA
| | - Sydney R. VanLinden
- Asthma and Airway Disease Research Center, The University
of Arizona, Tucson, AZ, 85721, USA
| | - Sean P. Malone
- Asthma and Airway Disease Research Center, The University
of Arizona, Tucson, AZ, 85721, USA
| | - Amir Abidov
- Medical Student Research Program, College of Medicine, The
University of Arizona, Tucson, AZ, 85721, USA
| | - Dayna Anderson
- Asthma and Airway Disease Research Center, The University
of Arizona, Tucson, AZ, 85721, USA
| | - Justyna Gozdz
- Asthma and Airway Disease Research Center, The University
of Arizona, Tucson, AZ, 85721, USA
| | - Avery DeVries
- Asthma and Airway Disease Research Center, The University
of Arizona, Tucson, AZ, 85721, USA,The BIO5 Institute, The University of Arizona, Tucson, AZ,
85721, USA
| | - Fernando D. Martinez
- Asthma and Airway Disease Research Center, The University
of Arizona, Tucson, AZ, 85721, USA,The BIO5 Institute, The University of Arizona, Tucson, AZ,
85721, USA
| | | | - Donata Vercelli
- Department of Cellular and Molecular Medicine, The
University of Arizona, Tucson, AZ, 85721, USA,Asthma and Airway Disease Research Center, The University
of Arizona, Tucson, AZ, 85721, USA,The BIO5 Institute, The University of Arizona, Tucson, AZ,
85721, USA,Arizona Center for the Biology of Complex Diseases, The
University of Arizona
| |
Collapse
|
14
|
Troy NM, Strickland D, Serralha M, de Jong E, Jones AC, Read J, Galbraith S, Islam Z, Kaur P, Mincham KT, Holt BJ, Sly PD, Bosco A, Holt PG. Protection against severe infant lower respiratory tract infections by immune training: Mechanistic studies. J Allergy Clin Immunol 2022; 150:93-103. [PMID: 35177255 DOI: 10.1016/j.jaci.2022.01.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 12/23/2021] [Accepted: 01/10/2022] [Indexed: 10/19/2022]
Abstract
BACKGROUND Results from recent clinical studies suggest potential efficacy of immune training (IT)-based approaches for protection against severe lower respiratory tract infections in infants, but underlying mechanisms are unclear. OBJECTIVE We used systems-level analyses to elucidate IT mechanisms in infants in a clinical trial setting. METHODS Pre- and posttreatment peripheral blood mononuclear cells from a placebo-controlled trial in which winter treatment with the IT agent OM85 reduced infant respiratory infection frequency and/or duration were stimulated for 24 hours with the virus/bacteria mimics polyinosinic:polycytidylic acid/lipopolysaccharide. Transcriptomic profiling via RNA sequencing, pathway and upstream regulator analyses, and systems-level gene coexpression network analyses were used sequentially to elucidate and compare responses in treatment and placebo groups. RESULTS In contrast to subtle changes in antivirus-associated polyinosinic:polycytidylic acid response profiles, the bacterial lipopolysaccharide-triggered gene coexpression network responses exhibited OM85 treatment-associated upregulation of IFN signaling. This was accompanied by network rewiring resulting in increased coordination of TLR4 expression with IFN pathway-associated genes (especially master regulator IRF7); segregation of TNF and IFN-γ (which potentially synergize to exaggerate inflammatory sequelae) into separate expression modules; and reduced size/complexity of the main proinflammatory network module (containing, eg, IL-1,IL-6, and CCL3). Finally, we observed a reduced capacity for lipopolysaccharide-induced inflammatory cytokine (eg, IL-6 and TNF) production in the OM85 group. CONCLUSION These changes are consistent with treatment-induced enhancement of bacterial pathogen detection/clearance capabilities concomitant with enhanced capacity to regulate ensuing inflammatory response intensity and duration. We posit that IT agents exemplified by OM85 potentially protect against severe lower respiratory tract infections in infants principally by effects on innate immune responses targeting the bacterial components of the mixed respiratory viral/bacterial infections that are characteristic of this age group.
Collapse
Affiliation(s)
- Niamh M Troy
- Telethon Kids Institute, The University of Western Australia, Perth, Australia
| | - Deborah Strickland
- Telethon Kids Institute, The University of Western Australia, Perth, Australia
| | - Michael Serralha
- Telethon Kids Institute, The University of Western Australia, Perth, Australia
| | - Emma de Jong
- Telethon Kids Institute, The University of Western Australia, Perth, Australia
| | - Anya C Jones
- Telethon Kids Institute, The University of Western Australia, Perth, Australia
| | - James Read
- Telethon Kids Institute, The University of Western Australia, Perth, Australia
| | - Sally Galbraith
- Child Health Research Centre, University of Queensland, Brisbane, Australia
| | - Zahir Islam
- Child Health Research Centre, University of Queensland, Brisbane, Australia
| | - Parwinder Kaur
- School of Agriculture and Environment, The University of Western Australia, Perth, Australia
| | - Kyle T Mincham
- National Hearth and Lung Institute, Imperial College London, London, United Kingdom
| | - Barbara J Holt
- Telethon Kids Institute, The University of Western Australia, Perth, Australia
| | - Peter D Sly
- Child Health Research Centre, University of Queensland, Brisbane, Australia
| | - Anthony Bosco
- Asthma and Airway Disease Research Center, The University of Arizona, Tucson
| | - Patrick G Holt
- Telethon Kids Institute, The University of Western Australia, Perth, Australia.
| |
Collapse
|
15
|
Sex differences in innate anti-viral immune responses to respiratory viruses and in their clinical outcomes in a birth cohort study. Sci Rep 2021; 11:23741. [PMID: 34887467 PMCID: PMC8660814 DOI: 10.1038/s41598-021-03044-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 11/23/2021] [Indexed: 11/21/2022] Open
Abstract
The mechanisms explaining excess morbidity and mortality in respiratory infections among males are poorly understood. Innate immune responses are critical in protection against respiratory virus infections. We hypothesised that innate immune responses to respiratory viruses may be deficient in males. We stimulated peripheral blood mononuclear cells from 345 participants at age 16 years in a population-based birth cohort with three live respiratory viruses (rhinoviruses A16 and A1, and respiratory syncytial virus) and two viral mimics (R848 and CpG-A, to mimic responses to SARS-CoV-2) and investigated sex differences in interferon (IFN) responses. IFN-α responses to all viruses and stimuli were 1.34-2.06-fold lower in males than females (P = 0.018 - < 0.001). IFN-β, IFN-γ and IFN-induced chemokines were also deficient in males across all stimuli/viruses. Healthcare records revealed 12.1% of males and 6.6% of females were hospitalized with respiratory infections in infancy (P = 0.017). In conclusion, impaired innate anti-viral immunity in males likely results in high male morbidity and mortality from respiratory virus infections.
Collapse
|
16
|
Holt P, Strickland D. Innate Immune Training for Prevention of Recurrent Wheeze in Early Childhood. Am J Respir Crit Care Med 2021; 204:392-394. [PMID: 33844949 PMCID: PMC8480254 DOI: 10.1164/rccm.202103-0698ed] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Patrick Holt
- Telethon Kids Institute The University of Western Australia Nedlands, Western Australia, Australia
| | - Deborah Strickland
- Telethon Kids Institute The University of Western Australia Nedlands, Western Australia, Australia
| |
Collapse
|
17
|
Martins Costa Gomes G, de Gouveia Belinelo P, Starkey MR, Murphy VE, Hansbro PM, Sly PD, Robinson PD, Karmaus W, Gibson PG, Mattes J, Collison AM. Cord blood group 2 innate lymphoid cells are associated with lung function at 6 weeks of age. Clin Transl Immunology 2021; 10:e1296. [PMID: 34306680 PMCID: PMC8292948 DOI: 10.1002/cti2.1296] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 04/27/2021] [Accepted: 05/21/2021] [Indexed: 11/21/2022] Open
Abstract
Objective Offspring born to mothers with asthma in pregnancy are known to have lower lung function which tracks with age. Human group 2 innate lymphoid cells (ILC2) accumulate in foetal lungs, at 10‐fold higher levels compared to adult lungs. However, there are no data on foetal ILC2 numbers and the association with respiratory health outcomes such as lung function in early life. We aimed to investigate cord blood immune cell populations from babies born to mothers with asthma in pregnancy. Methods Cord blood from babies born to asthmatic mothers was collected, and cells were stained in whole cord blood. Analyses were done using traditional gating approaches and computational methodologies (t‐distributed stochastic neighbour embedding and PhenoGraph algorithms). At 6 weeks of age, the time to peak tidal expiratory flow as a percentage of total expiratory flow time (tPTEF/tE%) was determined as well as Lung Clearance Index (LCI), during quiet natural sleep. Results Of 110 eligible infants (March 2017 to November 2019), 91 were successfully immunophenotyped (82.7%). Lung function was attempted in 61 infants (67.0%), and 43 of those infants (70.5% of attempted) had technically acceptable tPTEF/tE% measurements. Thirty‐four infants (55.7% of attempted) had acceptable LCI measurements. Foetal ILC2 numbers with increased expression of chemoattractant receptor‐homologous molecule (CRTh2), characterised by two distinct analysis methodologies, were associated with poorer infant lung function at 6 weeks of age.” Conclusion Foetal immune responses may be a surrogate variable for or directly influence lung function outcomes in early life.
Collapse
Affiliation(s)
- Gabriela Martins Costa Gomes
- Priority Research Centre GrowUpWell® - Hunter Medical Research Institute The University of Newcastle Newcastle NSW Australia
| | - Patricia de Gouveia Belinelo
- Priority Research Centre GrowUpWell® - Hunter Medical Research Institute The University of Newcastle Newcastle NSW Australia
| | - Malcolm R Starkey
- Priority Research Centre GrowUpWell® - Hunter Medical Research Institute The University of Newcastle Newcastle NSW Australia.,Priority Research Centre for Healthy Lungs - Hunter Medical Research Institute University of Newcastle Newcastle NSW Australia.,Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia
| | - Vanessa E Murphy
- Priority Research Centre GrowUpWell® - Hunter Medical Research Institute The University of Newcastle Newcastle NSW Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs - Hunter Medical Research Institute University of Newcastle Newcastle NSW Australia.,Centenary UTS Centre for Inflammation Centenary Institute Sydney NSW Australia
| | - Peter D Sly
- Child Health Research Centre University of Queensland Brisbane QLD Australia
| | - Paul D Robinson
- Department of Respiratory Medicine The Children's Hospital at Westmead Sydney NSW Australia
| | | | - Peter G Gibson
- Priority Research Centre for Healthy Lungs - Hunter Medical Research Institute University of Newcastle Newcastle NSW Australia.,Sleep Medicine Department John Hunter Hospital Newcastle NSW Australia
| | - Joerg Mattes
- Priority Research Centre GrowUpWell® - Hunter Medical Research Institute The University of Newcastle Newcastle NSW Australia.,Paediatric Respiratory & Sleep Medicine Department John Hunter Children's Hospital Newcastle NSW Australia
| | - Adam M Collison
- Priority Research Centre GrowUpWell® - Hunter Medical Research Institute The University of Newcastle Newcastle NSW Australia
| |
Collapse
|
18
|
Lauzon-Joset JF, Mincham KT, Scott NM, Khandan Y, Stumbles PA, Holt PG, Strickland DH. Protection against neonatal respiratory viral infection via maternal treatment during pregnancy with the benign immune training agent OM-85. Clin Transl Immunology 2021; 10:e1303. [PMID: 34249358 PMCID: PMC8248556 DOI: 10.1002/cti2.1303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/20/2021] [Accepted: 06/02/2021] [Indexed: 12/29/2022] Open
Abstract
Objectives Incomplete maturation of immune regulatory functions at birth is antecedent to the heightened risk for severe respiratory infections during infancy. Our forerunner animal model studies demonstrated that maternal treatment with the microbial‐derived immune training agent OM‐85 during pregnancy promotes accelerated postnatal maturation of mechanisms that regulate inflammatory processes in the offspring airways. Here, we aimed to provide proof of concept for a novel solution to reduce the burden and potential long‐term sequelae of severe early‐life respiratory viral infection through maternal oral treatment during pregnancy with OM‐85, already in widespread human clinical use. Methods In this study, we performed flow cytometry and targeted gene expression (RT‐qPCR) analysis on lungs from neonatal offspring whose mothers received oral OM‐85 treatment during pregnancy. We next determined whether neonatal offspring from OM‐85 treated mothers demonstrate enhanced protection against lethal lower respiratory infection with mouse‐adapted rhinovirus (vMC0), and associated lung immune changes. Results Offspring from mothers treated with OM‐85 during pregnancy display accelerated postnatal seeding of lung myeloid populations demonstrating upregulation of function‐associated markers. Offspring from OM‐85 mothers additionally exhibit enhanced expression of TLR4/7 and the IL‐1β/NLRP3 inflammasome complex within the lung. These treatment effects were associated with enhanced capacity to clear an otherwise lethal respiratory viral infection during the neonatal period, with concomitant regulation of viral‐induced IFN response intensity. Conclusion These results demonstrate that maternal OM‐85 treatment protects offspring against lethal neonatal respiratory viral infection by accelerating development of innate immune mechanisms crucial for maintenance of local immune homeostasis in the face of pathogen challenge.
Collapse
Affiliation(s)
- Jean-Francois Lauzon-Joset
- Centre de Recherche Institut Universitaire de Cardiologie et de Pneumologie de Québec Université Laval Québec QC Canada.,Telethon Kids Institute University of Western Australia Nedlands WA Australia
| | - Kyle T Mincham
- Telethon Kids Institute University of Western Australia Nedlands WA Australia
| | - Naomi M Scott
- Telethon Kids Institute University of Western Australia Nedlands WA Australia
| | - Yasmine Khandan
- Telethon Kids Institute University of Western Australia Nedlands WA Australia
| | - Philip A Stumbles
- Telethon Kids Institute University of Western Australia Nedlands WA Australia.,Medical, Molecular and Forensic Sciences Murdoch University Perth WA Australia
| | - Patrick G Holt
- Telethon Kids Institute University of Western Australia Nedlands WA Australia
| | | |
Collapse
|
19
|
Lynch SV, Vercelli D. Microbiota, Epigenetics, and Trained Immunity. Convergent Drivers and Mediators of the Asthma Trajectory from Pregnancy to Childhood. Am J Respir Crit Care Med 2021; 203:802-808. [PMID: 33493428 DOI: 10.1164/rccm.202010-3779pp] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Susan V Lynch
- Division of Gastroenterology, Department of Medicine, University of California, San Francisco, California; and
| | - Donata Vercelli
- Department of Cellular and Molecular Medicine & Asthma and Airway Disease Research Center, University of Arizona, Tucson, Arizona
| |
Collapse
|
20
|
Distinct Antiviral Properties of Two Different Bacterial Lysates. Can Respir J 2021; 2021:8826645. [PMID: 33613792 PMCID: PMC7878088 DOI: 10.1155/2021/8826645] [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: 09/11/2020] [Accepted: 01/20/2021] [Indexed: 12/23/2022] Open
Abstract
Oral bacterial lysates (OBLs) can reduce the frequency and severity of recurrent respiratory tract infections in children from viral and bacterial origins. OBL-induced early innate immune reaction was already shown, but the specific features of different OBLs have never been studied and compared. A study was conducted to assess in vitro the protective effects on rhinovirus- (RV-) infected human bronchial epithelial cells (BECs) of two slightly different OBLs: OM-85 and Pulmonarom. Furthermore, since immune cells represent the key arm for antiviral defence, the capacity of these OBLs to induce selected cytokine production in mouse bone marrow-derived DCs (BMDCs) was also evaluated. Although different OBLs may share some mechanisms to protect host cells from virus infection, some product-specific antimicrobial activities were observed on RV-infected human BECs and mouse BMDCs. These results are consistent with a product-specific response possibly triggered by different pathogen-associated molecular patterns (PAMPs) contained in OBLs.
Collapse
|
21
|
Pet ownership in pregnancy and methylation pattern in cord blood. Genes Immun 2021; 22:305-312. [PMID: 34642452 PMCID: PMC8506094 DOI: 10.1038/s41435-021-00151-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 09/13/2021] [Accepted: 09/28/2021] [Indexed: 11/09/2022]
Abstract
Having pets in the house during the first years of life has been shown to protect against allergies. However, the result of different studies is heterogeneous. The aim of this study was to evaluate the methylation pattern in cord blood in relation to pet ownership during pregnancy.We investigated the methylation patterns of 96 cord blood samples, participants of the Epigenetic Hallmark of Maternal Atopy and Diet-ELMA project, born to mothers who either owned pets (n = 32) or did not own pets (n = 64) during their pregnancy. DNA from cord blood was analysed using the Infinium methylation EPIC. For statistical analysis, RnBeads software was applied.We found 113 differentially methylated sites (DMs) in the covariate-adjusted analysis (FDR p < 0.05), with small methylation differences. The top DMs were associated with genes: UBA7, THRAP3, GTDC1, PDE8A and SBK2. In the regional analysis, two promoter regions presented with significance: RN7SL621P and RNU6-211P. Cis-regulatory element analysis revealed significant associations with several immune-related pathways, such as regulation of IL18, Toll signalling, IL6 and complement.We conclude that pet exposure during pregnancy causes subtle but significant changes in methylation patterns in cord blood, which are reflected in the biological processes governing both innate and adaptive immune responses.
Collapse
|
22
|
Mincham KT, Jones AC, Bodinier M, Scott NM, Lauzon-Joset JF, Stumbles PA, Bosco A, Holt PG, Strickland DH. Transplacental Innate Immune Training via Maternal Microbial Exposure: Role of XBP1-ERN1 Axis in Dendritic Cell Precursor Programming. Front Immunol 2020; 11:601494. [PMID: 33424847 PMCID: PMC7793790 DOI: 10.3389/fimmu.2020.601494] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 11/05/2020] [Indexed: 12/19/2022] Open
Abstract
We recently reported that offspring of mice treated during pregnancy with the microbial-derived immunomodulator OM-85 manifest striking resistance to allergic airways inflammation, and localized the potential treatment target to fetal conventional dendritic cell (cDC) progenitors. Here, we profile maternal OM-85 treatment-associated transcriptomic signatures in fetal bone marrow, and identify a series of immunometabolic pathways which provide essential metabolites for accelerated myelopoiesis. Additionally, the cDC progenitor compartment displayed treatment-associated activation of the XBP1-ERN1 signalling axis which has been shown to be crucial for tissue survival of cDC, particularly within the lungs. Our forerunner studies indicate uniquely rapid turnover of airway mucosal cDCs at baseline, with further large-scale upregulation of population dynamics during aeroallergen and/or pathogen challenge. We suggest that enhanced capacity for XBP1-ERN1-dependent cDC survival within the airway mucosal tissue microenvironment may be a crucial element of OM-85-mediated transplacental innate immune training which results in postnatal resistance to airway inflammatory disease.
Collapse
Affiliation(s)
- Kyle T. Mincham
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Anya C. Jones
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Marie Bodinier
- INRA Pays de la Loire, UR 1268 Biopolymers Interactions Assemblies (BIA) Nantes, Nantes, France
| | - Naomi M. Scott
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Jean-Francois Lauzon-Joset
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
- Centre de recherche de I‘Institut de Cardiologie et de Pneumologie de Québec, Université, Laval, QC, Canada
| | - Philip A. Stumbles
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
- College of Science, Health, Engineering and Education, Murdoch University, Perth, WA, Australia
| | - Anthony Bosco
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Patrick G. Holt
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
- Child Health Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | | |
Collapse
|
23
|
Decoding Susceptibility to Respiratory Viral Infections and Asthma Inception in Children. Int J Mol Sci 2020; 21:ijms21176372. [PMID: 32887352 PMCID: PMC7503410 DOI: 10.3390/ijms21176372] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 01/19/2023] Open
Abstract
Human Respiratory Syncytial Virus and Human Rhinovirus are the most frequent cause of respiratory tract infections in infants and children and are major triggers of acute viral bronchiolitis, wheezing and asthma exacerbations. Here, we will discuss the application of the powerful tools of systems biology to decode the molecular mechanisms that determine risk for infection and subsequent asthma. An important conceptual advance is the understanding that the innate immune system is governed by a Bow-tie architecture, where diverse input signals converge onto a few core pathways (e.g., IRF7), which in turn generate diverse outputs that orchestrate effector and regulatory functions. Molecular profiling studies in children with severe exacerbations of asthma/wheeze have identified two major immunological phenotypes. The IRF7hi phenotype is characterised by robust upregulation of antiviral response networks, and the IRF7lo phenotype is characterised by upregulation of markers of TGFβ signalling and type 2 inflammation. Similar phenotypes have been identified in infants and children with severe viral bronchiolitis. Notably, genome-wide association studies supported by experimental validation have identified key pathways that increase susceptibility to HRV infection (ORMDL3 and CHDR3) and modulate TGFβ signalling (GSDMB, TGFBR1, and SMAD3). Moreover, functional deficiencies in the activation of type I and III interferon responses are already evident at birth in children at risk of developing febrile lower respiratory tract infections and persistent asthma/wheeze, suggesting that the trajectory to asthma begins at birth or in utero. Finally, exposure to microbes and their products reprograms innate immunity and provides protection from the development of allergies and asthma in children, and therefore microbial products are logical candidates for the primary prevention of asthma.
Collapse
|