1
|
Ofri M, Kristal E, Cohen B, Beigelman A, Hazan G. The impact of neonatal antibiotic exposure on the development of childhood food allergies. Eur J Pediatr 2025; 184:304. [PMID: 40259094 PMCID: PMC12011889 DOI: 10.1007/s00431-025-06136-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/02/2025] [Accepted: 04/07/2025] [Indexed: 04/23/2025]
Abstract
Food allergies (FAs) in children have become increasingly prevalent. While early life factors such as gut microbiome disruptions have been implicated, the association between neonatal antibiotic exposure and subsequent FAs remains a topic of ongoing debate. This nationwide cohort study aimed to investigate the impact of neonatal antibiotic exposure on the development of childhood FA. This population-based retrospective cohort study analyzed data from Clalit-Healthcare-Services, Israel's largest state-mandated healthcare provider. The cohort included neonates (aged 0-60 days) admitted with fever between 2011 and 2018. Patients with confirmed infectious etiologies were excluded. The cohort was divided into two groups: those who received systemic antibiotics (Antibiotic ( +)) and those who did not (Antibiotic ( -)). FA cases were identified using ICD-9 codes up to age 6. Multivariate logistic regression and survival analysis models were utilized and adjusted for inflammatory markers, maternal atopy, and socioeconomic status. Among 2780 neonates, 1220 received antibiotics, while 1560 did not. The incidence of FAs was significantly higher in the Antibiotic ( +) group compared to the Antibiotic ( -) group (2.5% vs. 1.3%, P = 0.02). Adjusted analysis revealed that systemic antibiotic exposure during the neonatal period was associated with a threefold increased risk of FA up to age 6 (OR = 2.89, 95% CI = 1.34-6.92, P = 0.01). Conclusions: This study provides strong evidence linking neonatal antibiotic exposure to an increased risk of childhood FAs, particularly in the first 2 years of life. The findings highlight the importance of judicious antibiotic use in young infants.
Collapse
Affiliation(s)
- Mai Ofri
- The School of Medicine, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Eyal Kristal
- The Kipper Institute of Allergy and Immunology, Schneider Children's Medical Center of Israel, Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
- The Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Braha Cohen
- Clinical Research Center, Soroka University Medical Center, Beer Sheva, Israel
| | - Avraham Beigelman
- The Kipper Institute of Allergy and Immunology, Schneider Children's Medical Center of Israel, Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Guy Hazan
- The Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
- Pediatric Pulmonary Unit, Saban Children's Hospital, Soroka University Medical Center, Beer Sheva, Israel.
| |
Collapse
|
2
|
Badolati I, de Jong Y, Uhl C, Ullberg J, Joustra M, Fagerberg UL, Nilsson C, Asarnoj A, Sverremark‐Ekström E. One Year of Oral Immunotherapy Impacts the Gut Microbiota and Plasma Metabolome of Peanut-Allergic Young Children. Clin Exp Allergy 2025; 55:340-343. [PMID: 39602883 PMCID: PMC11994247 DOI: 10.1111/cea.14607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024]
Affiliation(s)
- Isabella Badolati
- Department of Molecular Biosciences, The Wenner‐Gren InstituteStockholm UniversityStockholmSweden
| | - Ymke de Jong
- Department of Molecular Biosciences, The Wenner‐Gren InstituteStockholm UniversityStockholmSweden
| | - Carina Uhl
- Department of Clinical Science and Education, SödersjukhusetKarolinska InstitutetStockholmSweden
- Sachs' Children and Youth HospitalStockholmSweden
| | - Josefin Ullberg
- Department of PediatricsVästmanland HospitalVästeråsSweden
- Centre for Innovation, Research and Education, Region VästmanlandVästmanland HospitalVästeråsSweden
| | - Marleen Joustra
- Department of Molecular Biosciences, The Wenner‐Gren InstituteStockholm UniversityStockholmSweden
| | - Ulrika Lorentzon Fagerberg
- Department of PediatricsVästmanland HospitalVästeråsSweden
- Centre for Innovation, Research and Education, Region VästmanlandVästmanland HospitalVästeråsSweden
- Department of Women's and Children's HealthKarolinska InstitutetStockholmSweden
| | - Caroline Nilsson
- Department of Clinical Science and Education, SödersjukhusetKarolinska InstitutetStockholmSweden
- Sachs' Children and Youth HospitalStockholmSweden
| | - Anna Asarnoj
- Department of Women's and Children's HealthKarolinska InstitutetStockholmSweden
- Pediatric Allergy and Pulmonology Unit at Astrid Lindgren Children's HospitalKarolinska University HospitalStockholmSweden
| | - Eva Sverremark‐Ekström
- Department of Molecular Biosciences, The Wenner‐Gren InstituteStockholm UniversityStockholmSweden
| |
Collapse
|
3
|
Wang Y, Yang J, Lai X, Leung ASY, Xing Y, Wong GWK. Early Origins of Asthma and Allergies: Clues From Studies in China. Clin Exp Allergy 2025. [PMID: 40087850 DOI: 10.1111/cea.70033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 02/28/2025] [Accepted: 03/01/2025] [Indexed: 03/17/2025]
Abstract
Asthma and allergies have emerged as some of the most common chronic diseases, particularly in developed countries. Epidemiological studies have consistently demonstrated that children growing up in farming/rural environments are less likely to develop these conditions. Over the past three decades, China has experienced unprecedented economic development and urbanisation, accompanied by a rapid rise in the prevalence of allergic disorders. Despite the substantial number of affected individuals, allergy management in China remains inconsistent and often inadequate, compounded by variations in diagnostic criteria and limited healthcare access in less developed regions. Furthermore, the vast population, regional disparities, and methodological inconsistencies in data collection have hindered the acquisition of comprehensive, large-scale epidemiological data. This review examines the factors contributing to asthma and allergies from their early origins, focusing on modifiable factors from a specific perspective of China. Factors related to traditional lifestyle, such as early-life exposure to agricultural farming and poultry, diverse dietary patterns, and early introduction of allergenic foods, appear to offer protection against allergies. Conversely, exposure to open-fire cooking, incense burning, tobacco smoke, as well as early-life antibiotic use and perinatal factors like Caesarean section delivery and prematurity may represent potential risks. A clear understanding of the role of these factors would pave the way for developing effective interventions to mitigate the substantial health and socioeconomic burdens associated with asthma and allergies.
Collapse
Affiliation(s)
- Yike Wang
- School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, China
| | - Jing Yang
- Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Xueling Lai
- Shenzhen Guangming Maternal & Child Healthcare Hospital, Shenzhen, China
| | - Agnes Sze-Yin Leung
- Department of Paediatrics, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Yuhan Xing
- School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, China
- Department of Paediatrics, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Gary Wing-Kin Wong
- Department of Paediatrics, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, SAR, China
| |
Collapse
|
4
|
Jung M, Lee JY, Kim S, Song J, Jang S, Shin S, Lee MH, Kim MJ, Kim J, Lee HB, Kim Y, Ahn K, Kim M, Kim J. Altered diversity and composition of gut microbiota in Korean children with food allergy. Clin Transl Allergy 2025; 15:e70036. [PMID: 40074707 PMCID: PMC11903216 DOI: 10.1002/clt2.70036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/20/2024] [Accepted: 01/20/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND This study aimed to comprehensively characterize the gut microbiome and identify individual and grouped gut microbes associated with food allergy (FA) using 16S rRNA gene sequencing. METHODS Fecal samples were collected from children with IgE-mediated FA and from sex- and age-matched controls. The V3-V4 variable regions of the 16S rRNA gene of the gut microbiome were profiled using next-generation sequencing (Illumina, USA). Bacterial species richness, intracommunity diversity, and intergroup dissimilarity were evaluated. Functional profiles were predicted using Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt) and the Minimal Set of Pathways (MinPath) algorithm. RESULTS Fecal samples were collected from children with IgE-mediated FA (n = 66) and from sex- and age-matched controls (n = 22). Gut microbiome richness (p < 0.0001), intra-community diversity (p < 0.0001), and inter-community diversity (p = 0.0004) were higher in the healthy group than in the FA group. Patients with FA were enriched in Blautia, Fusicatenibacter, and Ruminococcus_g5 compared with healthy control individuals (all p < 0.05). Healthy control individuals were significantly enriched in Oscillibacter and Ruminococcus compared with patients with FA (all p < 0.05). Functional pathway analysis identified enrichment in pathways related to endoglucanase in healthy controls and the ATP-binding cassette (ABC) transport system in FA patients. CONCLUSIONS The gut microbiomes of patients with FA and healthy control individuals had different taxonomic abundances, and the microbiome richness and diversity of the bacterial flora of patients with FA were reduced compared with controls.
Collapse
Affiliation(s)
- Minyoung Jung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Ji Young Lee
- Department of Pediatrics, Hallym University Chuncheon Sacred Heart Hospital, Chuncheon, South Korea
| | - Sukyung Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jeongmin Song
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Sehun Jang
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Sanghee Shin
- Department of Pediatrics, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, South Korea
| | - Min Hee Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Mi Jin Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jiwon Kim
- Department of Pediatrics, Chungnam National University Sejong Hospital, Sejong, South Korea
| | - Han Byul Lee
- Research Institute for Future Medical Science, Chungnam National University Sejong Hospital, Sejong, South Korea
| | - Yeonghee Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Kangmo Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Seoul, South Korea
| | - Minji Kim
- Department of Pediatrics, Chungnam National University Sejong Hospital, Sejong, South Korea
| | - Jihyun Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Seoul, South Korea
| |
Collapse
|
5
|
Nowak-Wegrzyn A, Sicherer SH, Akin C, Anvari S, Bartnikas LM, Berin MC, Bingemann TA, Boyd S, Brown-Whitehorn T, Bunyavanich S, Cianferoni A, du Toit G, Fortunato JE, Goldsmith JD, Groetch M, Leonard SA, Rao M, Schultz F, Schwaninger JM, Venter C, Westcott-Chavez A, Wood RA, Togias A. Current status and future directions in food protein-induced enterocolitis syndrome: An NIAID workshop report of the June 22, 2022, virtual meeting. J Allergy Clin Immunol 2025; 155:336-356. [PMID: 39521282 DOI: 10.1016/j.jaci.2024.10.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/10/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024]
Abstract
Food protein-induced enterocolitis syndrome (FPIES) is a non-IgE-mediated gastrointestinal food allergy characterized by delayed, protracted vomiting and accompanied by lethargy and pallor, usually 1 to 4 hours after ingesting the food allergen. The pathophysiology of FPIES remains unknown, and currently there are no diagnostic biomarkers available to assess disease activity or its resolution. Over the last 2 decades, FPIES has become increasingly recognized in both pediatric and adult patients. Forty years after the initial FPIES description, the first FPIES code appeared in the International Classification of Diseases, Tenth Revision (ICD-10), and the first international consensus guidelines for the diagnosis and management of FPIES were published. On June 22, 2022, the National Institute of Allergy and Infectious Diseases (NIAID) held its first virtual multidisciplinary workshop on FPIES. Various clinical and translational aspects of FPIES as well as important areas of unmet needs were discussed as priorities for future research during this 2-day virtual workshop. Our report provides a summary of content of the workshop, including updated literature on the topic areas, and also provides critical commentary on the state of FPIES.
Collapse
Affiliation(s)
- Anna Nowak-Wegrzyn
- Department of Pediatrics, Hassenfeld Children's Hospital, NYU Grossman School of Medicine, New York, and the Department of Pediatrics, Gastroenterology and Nutrition, Collegium Medicum, University of Warmia and Mazury, Olsztyn, Poland; Department of Pediatrics, Division of Pediatric Allergy and Immunology, Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, New York, NY.
| | - Scott H Sicherer
- Department of Pediatrics, Division of Pediatric Allergy and Immunology, Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Cem Akin
- Department of Medicine, University of Michigan, Division of Allergy and Clinical Immunology, Ann Arbor, Mich
| | - Sara Anvari
- Division of Immunology, Allergy and Retrovirology, Texas Children's Hospital, Baylor College of Medicine, Houston, and Texas Children's Hospital, William T. Shearer Center for Human Immunobiology, Houston, Tex
| | - Lisa M Bartnikas
- Department of Medicine, Division of Immunology, Boston Children's Hospital, Boston, and Harvard Medical School, Boston, Mass
| | - M Cecilia Berin
- Department of Medicine, Division of Allergy/Immunology, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Theresa A Bingemann
- Department of Allergy, Immunology and Rheumatology and the Department of Pediatric Allergy and Immunology, University of Rochester School of Medicine, Rochester, NY
| | - Scott Boyd
- Stanford University School of Medicine, Palo Alto, Calif
| | - Terri Brown-Whitehorn
- Division of Pediatric Allergy and Immunology, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, Pa
| | - Supinda Bunyavanich
- Department of Pediatrics, Division of Pediatric Allergy and Immunology, Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Antonella Cianferoni
- Division of Pediatric Allergy and Immunology, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, Pa
| | - George du Toit
- Department of Women and Children's Health (Paediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, and the Children's Allergy Service, Evelina London Children's Hospital, Guy's and St Thomas' Hospital, London, United Kingdom
| | - John E Fortunato
- Gastroenterology, Hepatology and Nutrition, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Jeffrey D Goldsmith
- Department of Pathology, Boston Children's Hospital, Boston, and Harvard Medical School, Boston, Mass
| | - Marion Groetch
- Department of Pediatrics, Division of Pediatric Allergy and Immunology, Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Stephanie A Leonard
- Division of Pediatric Allergy & Immunology, Rady Children's Hospital San Diego, University of California, San Diego, Calif
| | - Meenakshi Rao
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, and Harvard Medical School, Boston, Mass
| | - Fallon Schultz
- Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Rockville, Md
| | | | - Carina Venter
- Children's Hospital Colorado, University of Colorado, Denver, Colo
| | | | - Robert A Wood
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Alkis Togias
- Allergy, Immunology and Transplantation, NIAID, National Institutes of Health, Rockville, Md
| |
Collapse
|
6
|
Crabtree D, Seidler K, Barrow M. Pathophysiological mechanisms of gut dysbiosis and food allergy and an investigation of probiotics as an intervention for atopic disease. Clin Nutr ESPEN 2025; 65:189-204. [PMID: 39571752 DOI: 10.1016/j.clnesp.2024.11.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 11/15/2024] [Indexed: 12/08/2024]
Abstract
BACKGROUND AND AIMS Epidemiological studies have associated reduced bacterial diversity and abundance and food allergy. This mechanistic review investigated the link between gut dysbiosis and food allergy with a focus on the role of short-chain fatty acids (SCFAs) in modulating T-cells. T-cell differentiation poses an opportunity to direct the immune cells towards an anergic regulatory T cell (Treg) or allergic T helper 2 (Th2) response. Probiotic intervention to prevent and/or treat atopic disease symptoms through this mechanistic pathway was explored. METHODOLOGY A narrative review was conducted following a three-stage systematic literature search of EMBASE and Medline databases. Ninety-six of 571 papers were accepted and critically appraised using ARRIVE and SIGN50 forms. Thematic analysis identified key pathophysiological mechanisms within the narrative of included papers. RESULTS Preclinical studies provided compelling evidence for SCFAs' modulation of T-cell differentiation, which may act through G-protein coupled receptors 41, 43 and 109a and histone deacetylase inhibition. Foxp3 transcription factor was implicated in the upregulation of Tregs. Human probiotic intervention studies aimed at increasing SCFAs and Tregs and preventing atopic disease showed inconclusive results. However, evidence for probiotic intervention in children with cow's milk protein allergy (CMPA) was more promising and warrants further investigation. CONCLUSION Preclinical evidence suggests that the mechanism of gut dysbiosis and reduced SCFAs may skew T-cell differentiation towards a Th2 response, thus inducing allergy symptoms. Probiotic trials were inconclusive: probiotics were predominantly unsuccessful in the prevention of allergic disease, however, may be able to modulate food allergy symptoms in infants with CMPA.
Collapse
Affiliation(s)
- Danielle Crabtree
- Centre for Nutrition Education and Lifestyle Management, PO Box 3739, Wokingham, RG40 9UA, UK.
| | - Karin Seidler
- Centre for Nutrition Education and Lifestyle Management, PO Box 3739, Wokingham, RG40 9UA, UK.
| | - Michelle Barrow
- Centre for Nutrition Education and Lifestyle Management, PO Box 3739, Wokingham, RG40 9UA, UK.
| |
Collapse
|
7
|
Zhang L, Chun Y, Grishina G, Lo T, Reed K, Wang J, Sicherer S, Berin MC, Bunyavanich S. Oral and Gut Microbial Hubs Associated With Reaction Threshold Interact With Circulating Immune Factors in Peanut Allergy. Allergy 2025. [PMID: 39887792 DOI: 10.1111/all.16481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/09/2024] [Accepted: 12/19/2024] [Indexed: 02/01/2025]
Abstract
BACKGROUND Among peanut-allergic individuals, there is high variability in the amount of peanut that triggers reactions (i.e., reaction threshold) that is not predictable or well-understood. We conducted this study to characterize relationships between the oral and gut microbiomes and systemic processes associated with reaction threshold in peanut allergy (PA). METHODS In a cohort of 120 children with suspected PA who underwent double-blind, placebo-controlled food challenges, we generated and analyzed parallel profiles of the oral microbiome, gut microbiome, peripheral blood transcriptome, peripheral blood cytometry, and serum antibody levels to identify threshold-associated markers and their inter-relationships. RESULTS The 120 participants included 23 children with no PA, 74 with high-threshold PA (reacting to ≥ 443 mg cumulative peanut protein), and 23 with low-threshold PA (reacting to < 443 mg cumulative peanut protein). Ten hub microbes were each identified in saliva and stool microbiome networks that were constructed, including the hub microbes Rothia aeria in saliva and Bacteroides sp. in stool that were associated with reaction threshold. These hub microbes were also associated with peripheral blood transcript levels for threshold-associated key drivers of FcγR-mediated phagocytosis and TLR signaling. Correlation network construction with additional data on threshold-associated peripheral blood neutrophil abundance and peanut-specific serum IgE and Ara h 2 antibody levels revealed central roles for saliva Rothia aeria and stool Bacteroides sp. in local-systemic networks for IgE- and IgG-mediated peanut allergy. CONCLUSIONS This integrated study of oral and stool microbiomes, blood transcriptome, cellular profiles, and peanut-specific serum antibodies revealed new relationships between local microbiota and systemic measures associated with reaction threshold in peanut allergy.
Collapse
Affiliation(s)
- Lingdi Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Yoojin Chun
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Galina Grishina
- Division of Allergy and Immunology, Department of Pediatrics, Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Tracy Lo
- Division of Allergy and Immunology, Department of Pediatrics, Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kyle Reed
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Julie Wang
- Division of Allergy and Immunology, Department of Pediatrics, Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Scott Sicherer
- Division of Allergy and Immunology, Department of Pediatrics, Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - M Cecilia Berin
- Division of Allergy and Immunology, Department of Medicine, Northwestern Feinberg School of Medicine, Chicago, Illinois, USA
| | - Supinda Bunyavanich
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Division of Allergy and Immunology, Department of Pediatrics, Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
8
|
Lin X, Chen Y, Lin W. Bibliometric Analysis of Global Pediatric Research on Cow's Milk Protein Allergy. J Asthma Allergy 2025; 18:85-100. [PMID: 39872156 PMCID: PMC11771186 DOI: 10.2147/jaa.s487698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 01/08/2025] [Indexed: 01/29/2025] Open
Abstract
Background Cow's milk protein allergy (CMPA) is a prevalent food allergy in early childhood, significantly impacting the quality of life for affected children. Current palliative measures, such as specialized formula milk, offer temporary relief but are costly and fail to address the underlying issue. Thus, there is a critical need to better understand CMPA and explore new treatment options. Methods This study employed bibliometric methods to analyze global pediatric CMPA research and identify future directions for the first time. Visual analyses were conducted using VOS Viewer and CiteSpace software. Results A total of 2040 articles published between 2000 and 2023 showed increasing annual publications. In this field of research, the Icahn School of Medicine at Mount Sinai has made significant contributions, with the most influential articles published in the Journal of Allergy and Clinical Immunology. Current research emphasizes personalized therapy, probiotics, and gut microbiota in CMPA. Conclusion Future research will focus on microbiota-related personalized treatments, promising effective clinical interventions.
Collapse
Affiliation(s)
- Xiaolan Lin
- Department of Pediatric Gastroenterology, Fuzhou Children’s Hospital of Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
| | - Yifan Chen
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
- Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Fuzhou, Fujian, People’s Republic of China
| | - Weidong Lin
- Department of Pediatric Gastroenterology, Fuzhou Children’s Hospital of Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
| |
Collapse
|
9
|
Bodén S, Lindam A, Venter C, Ulfsdotter RL, Domellöf M, West CE. Diversity of complementary diet and early food allergy risk. Pediatr Allergy Immunol 2025; 36:e70035. [PMID: 39868464 PMCID: PMC11771557 DOI: 10.1111/pai.70035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/17/2024] [Accepted: 01/16/2025] [Indexed: 01/28/2025]
Abstract
INTRODUCTION Diet diversity (DD) in infancy may be protective for early food allergy (FA) but there is limited knowledge about how DD incorporating consumption frequency influences FA risk. METHODS Three measures of DD were investigated in 2060 infants at 6 and/or at 9 months of age within the NorthPop Birth Cohort Study: a weighted DD score based on intake frequency, the number of introduced foods, and the number of introduced allergenic foods. In multivariable logistic regression models based on directed acyclic graphs, associations to parentally reported physician-diagnosed FA at age 9 and 18 months were estimated, including sensitivity and stratified analyses. RESULTS High weighted DD scores (24-31p) at age 9 months were associated with 61% decreased odds of FA at age 18 months [OR (95% CI) = 0.39 0.18-0.88] compared with infants with the lowest DD scores (0-17p). The association remained significant after exclusion of early FA cases. Having introduced 13-14 foods at age 9 months, independent of consumption frequency, was associated with 45% decreased odds of FA [OR (95% CI) = 0.55 (0.31-0.98)] compared to having introduced 0-10 foods. When stratifying, significantly reduced odds for FA were seen for children with eczema and for children with no FA history in the family. No association was seen between DD at age 6 months and FA at age 18 months. CONCLUSION A diverse diet at age 9 months may prevent FA at age 18 months. Our results underscore the need for additional investigations on the impact of consumption frequency in infancy.
Collapse
Affiliation(s)
- Stina Bodén
- Department of Clinical Sciences, PediatricsUmeå UniversityUmeåSweden
| | - Anna Lindam
- Department of Public Health and Clinical Medicine, Unit of Research, Education and Development – ÖstersundUmeå UniversityUmeåSweden
| | - Carina Venter
- Section of Allergy & Immunology, Department of Pediatrics, Children's Hospital ColoradoUniversity of Colorado School of MedicineAuroraColoradoUSA
- Children's Hospital ColoradoAuroraColoradoUSA
| | | | - Magnus Domellöf
- Department of Clinical Sciences, PediatricsUmeå UniversityUmeåSweden
| | - Christina E. West
- Department of Clinical Sciences, PediatricsUmeå UniversityUmeåSweden
| |
Collapse
|
10
|
Ma J, Palmer DJ, Geddes D, Lai CT, Rea A, Prescott SL, D'Vaz N, Stinson LF. Maternal Allergic Disease Phenotype and Infant Birth Season Influence the Human Milk Microbiome. Allergy 2024. [PMID: 39723602 DOI: 10.1111/all.16442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 10/11/2024] [Accepted: 11/19/2024] [Indexed: 12/28/2024]
Abstract
Early infancy is a critical period for immune development. In addition to being the primary food source during early infancy, human milk also provides multiple bioactive components that shape the infant gut microbiome and immune system and provides a constant source of exposure to maternal microbiota. Given the potential interplay between allergic diseases and the human microbiome, this study aimed to characterise the milk microbiome of allergic mothers. Full-length 16S rRNA gene sequencing was performed on milk samples collected at 3 and 6 months postpartum from 196 women with allergic disease. Multivariate linear mixed models were constructed to identify the maternal, infant, and environmental determinants of the milk microbiome. Human milk microbiome composition and beta diversity varied over time (PERMANOVA R2 = 0.011, p = 0.011). The season of infant birth emerged as the strongest determinant of the microbiome community structure (PERMANOVA R2 = 0.014, p = 0.011) with impacts on five of the most abundant taxa. The milk microbiome also varied according to the type of maternal allergic disease (allergic rhinitis, asthma, atopic dermatitis, and food allergy). Additionally, infant formula exposure reduced the relative abundance of several typical oral taxa in milk. In conclusion, the milk microbiome of allergic mothers was strongly shaped by the season of infant birth, maternal allergic disease phenotype, and infant feeding mode. Maternal allergic disease history and infant season of birth should therefore be considered in future studies of infant and maternal microbiota. Trial Registration: ClinicalTrials.gov identifier: ACTRN12606000281594.
Collapse
Affiliation(s)
- Jie Ma
- School of Molecular Sciences, The University of Western Australia, Crawley, WA, Australia
- ABREAST Network, Perth, WA, Australia
- UWA Centre for Human Lactation Research and Translation, Perth, WA, Australia
| | - Debra J Palmer
- ABREAST Network, Perth, WA, Australia
- Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
- School of Medicine, The University of Western Australia, Perth, WA, Australia
| | - Donna Geddes
- School of Molecular Sciences, The University of Western Australia, Crawley, WA, Australia
- ABREAST Network, Perth, WA, Australia
- UWA Centre for Human Lactation Research and Translation, Perth, WA, Australia
| | - Ching Tat Lai
- School of Molecular Sciences, The University of Western Australia, Crawley, WA, Australia
- ABREAST Network, Perth, WA, Australia
- UWA Centre for Human Lactation Research and Translation, Perth, WA, Australia
| | - Alethea Rea
- Mathematics and Statistics, Harry Butler Institute, Murdoch University, Murdoch, WA, Australia
| | - Susan L Prescott
- Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
- School of Medicine, The University of Western Australia, Perth, WA, Australia
- Nova Institute for Health, Baltimore, Maryland, USA
- Department of Family and Community Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Nina D'Vaz
- Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
- School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia
| | - Lisa F Stinson
- School of Molecular Sciences, The University of Western Australia, Crawley, WA, Australia
- ABREAST Network, Perth, WA, Australia
- UWA Centre for Human Lactation Research and Translation, Perth, WA, Australia
| |
Collapse
|
11
|
Todorov SD, Tagg J, Algburi A, Tiwari SK, Popov I, Weeks R, Mitrokhin OV, Kudryashov IA, Kraskevich DA, Chikindas ML. The Hygienic Significance of Microbiota and Probiotics for Human Wellbeing. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10419-9. [PMID: 39688648 DOI: 10.1007/s12602-024-10419-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2024] [Indexed: 12/18/2024]
Abstract
The human body can be viewed as a combination of ecological niches inhabited by trillions of bacteria, viruses, fungi, and parasites, all united by the microbiota concept. Human health largely depends on the nature of these relationships and how they are built and maintained. However, personal hygiene practices have historically been focused on the wholesale elimination of pathogens and "hygiene-challenging microorganisms" without considering the collateral damage to beneficial and commensal species. The microbiota can vary significantly in terms of the qualitative and quantitative composition both between different people and within one person during life, and the influence of various environmental factors, including age, nutrition, bad habits, genetic factors, physical activity, medication, and hygienic practices, facilitates these changes. Disturbance of the microbiota is a predisposing factor for the development of diseases and also greatly influences the course and severity of potential complications. Therefore, studying the composition of the microbiota of the different body systems and its appropriate correction is an urgent problem in the modern world. The application of personal hygiene products or probiotics must not compromise health through disruption of the healthy microbiota. Where changes in the composition or metabolic functions of the microbiome may occur, they must be carefully evaluated to ensure that essential biological functions are unaffected. As such, the purpose of this review is to consider the microbiota of each of the "ecological niches" of the human body and highlight the importance of the microbiota in maintaining a healthy body as well as the possibility of its modulation through the use of probiotics for the prevention and treatment of certain human diseases.
Collapse
Affiliation(s)
- Svetoslav D Todorov
- ProBacLab, Laboratório de Microbiologia de Alimentos, Departamento de Alimentos E Nutrição Experimental, Food Research Center, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, SP, 05508-000, Brazil.
- Instituto Politécnico de Viana Do Castelo, 4900-347, Viana Do Castelo, Portugal.
| | - John Tagg
- Blis Technologies, South Dunedin, 9012, New Zealand
| | - Ammar Algburi
- Department of Microbiology, College of Veterinary Medicine, University of Diyala, Baqubah, Iraq
| | - Santosh Kumar Tiwari
- Department of Genetics, Maharshi Dayanand University, Rohtak, 124001, Haryana, India
| | - Igor Popov
- Center for Agrobiotechnology, Don State Technical University, Gagarina Sq., 1344002, Rostov-On-Don, Russia
- Division of Immunobiology and Biomedicine, Center of Genetics and Life Sciences, Sirius University of Science and Technology, Federal Territory Sirius, Olimpijskij Ave., 1, Federal Territory Sirius, Sirius, 354340, Russia
- Centre for Healthy Eating & Food Innovation (HEFI), Maastricht University-Campus Venlo, Villafloraweg, 1, 5928 SZ, Venlo, The Netherlands
| | - Richard Weeks
- Health Promoting Naturals Laboratory, School of Environmental and Biological Sciences, Rutgers, the State University of New Jersey, New Brunswick, NJ, 08901, USA
| | - Oleg V Mitrokhin
- Department of General Hygiene, I.M. Sechenov First Moscow State Medical University, Moscow, 119435, Russia
| | - Ilya A Kudryashov
- Department of General Hygiene, I.M. Sechenov First Moscow State Medical University, Moscow, 119435, Russia
| | - Denis A Kraskevich
- Department of General Hygiene, I.M. Sechenov First Moscow State Medical University, Moscow, 119435, Russia
| | - Michael L Chikindas
- Center for Agrobiotechnology, Don State Technical University, Gagarina Sq., 1344002, Rostov-On-Don, Russia.
- Health Promoting Naturals Laboratory, School of Environmental and Biological Sciences, Rutgers, the State University of New Jersey, New Brunswick, NJ, 08901, USA.
- Department of General Hygiene, I.M. Sechenov First Moscow State Medical University, Moscow, 119435, Russia.
| |
Collapse
|
12
|
Steigerwald H, Albrecht M, Blissenbach B, Krause M, Wangorsch A, Schott M, Gonzalez-Menendez I, Quintanilla-Martinez L, Toda M, Vieths S, Krut O, Scheurer S, Blanco-Pérez F. Dietary fiber pectin alters the gut microbiota and diminishes the inflammatory immune responses in an experimental peach allergy mouse model. Sci Rep 2024; 14:30503. [PMID: 39681664 DOI: 10.1038/s41598-024-82210-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Since therapeutic options are limited the utilization of prebiotics is suggested to prevent food allergies (FAs). Using an experimental peach allergy model we explored the effect of dietary fiber pectin, a high-methoxyl heteropolysaccharide, on the manifestation of FA. CBA/J mice were sensitized, subsequently orally boosted and provoked with peach peel extract. For dietary intervention, mice were fed a pectin containing diet before (primary-preventive) or after (secondary-preventive) sensitization. Non-treated allergic and sham-treated mice were fed a diet containing 20% cellulose. Fecal microbiota, humoral and intestinal immune cell responses were analyzed. Pectin remarkably affected the gut microbiota composition and diversity, promoting mainly the growth of Bacteroides. The frequency of mast cells, macrophages, and CD3+T cells in the lamina propria of the small intestine was reduced, whereas the frequency of B cells and CD4+T cell subpopulation was enhanced. Pectin intervention in the primary-preventive stetting significantly triggered serum IgA levels, whereas production of IgE and mMCPT-1 was reduced. Remarkably, in both settings peach allergen-specific IgG1/IgG2a ratio and specific IgE were significantly reduced to baseline. The data suggest, that dietary supplementation of pectin in both intervention approaches can diminish inflammatory responses and signs of allergic immune responses, accompanied by alteration of the gut microbiota composition.
Collapse
Affiliation(s)
- Hanna Steigerwald
- Molecular Allergology, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | - Melanie Albrecht
- Molecular Allergology, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | - Birgit Blissenbach
- Microbiological Safety, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | - Maren Krause
- Molecular Allergology, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | - Andrea Wangorsch
- Molecular Allergology, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | - Maike Schott
- Molecular Allergology, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | - Irene Gonzalez-Menendez
- Institute of Pathology and Neuropathology, Comprehensive Cancer Center, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard-Karls University of Tübingen, Tübingen, Germany
| | - Leticia Quintanilla-Martinez
- Institute of Pathology and Neuropathology, Comprehensive Cancer Center, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard-Karls University of Tübingen, Tübingen, Germany
| | - Masako Toda
- Laboratory of Food and Biomolecular Science, Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, 980-8577, Japan
| | - Stefan Vieths
- Molecular Allergology, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | - Oleg Krut
- Microbiological Safety, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | - Stephan Scheurer
- Molecular Allergology, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen, Germany.
| | - Frank Blanco-Pérez
- Molecular Allergology, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| |
Collapse
|
13
|
Liu F, Zhang H, Fan L, Yu Q, Wang S. Hotspots and development trends of gut microbiota in atopic dermatitis: A bibliometric analysis from 1988 to 2024. Medicine (Baltimore) 2024; 103:e40931. [PMID: 39686442 DOI: 10.1097/md.0000000000040931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Atopic dermatitis (AD) is a prevalent inflammatory skin condition that commonly occurs in children. More and more scientific evidence suggests that gut microbiota plays an important role in the pathogenesis of AD, whereas there is no article providing a comprehensive summary and analysis. We aimed to analyze documents on AD and gut microbiota and identify hotspots and development trends in this field. METHODS Articles and reviews in the field of AD and gut microbiota from January 1, 1988 to October 20, 2024 were obtained from the Web of Science Core Collection database. Biblioshiny was utilized for evaluating and visualizing the core authors, journals, countries, documents, trend topics, and hotspots in this field. RESULTS Among 1672 documents, it indicated that the number of annual publications generally increased. The United States had the highest production, impact, and international collaboration. Journal of Allergy and Clinical Immunology was the journal of the maximum publications. Based on keyword co-occurrence and clustering analysis, "stratum-corneum lipids," "probiotics," "prebiotics," "fecal microbiota transplantation," "phage therapy," "short chain fatty-acids," "biologic therapy," and "skin inflammation" represented current trend topics. The pathological and molecular mechanisms and associated therapeutic methods for AD and gut microbiota were the research hotspots. The incorporation of microbiota-based therapies alongside conventional treatments can contribute to better clinical outcomes. CONCLUSION We highlighted that gut microbiota may exacerbate symptoms of AD through various aspects, including immunity, metabolites, and neuroendocrine pathways. More efforts are required to investigate the safety and efficacy of gut microbial management methods for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Fang Liu
- Hangzhou Linping District Maternal and Child Health Care Hospital, Hangzhou, China
| | - Haipeng Zhang
- Hangzhou Linping District Maternal and Child Health Care Hospital, Hangzhou, China
| | - Lina Fan
- Hangzhou Linping District Maternal and Child Health Care Hospital, Hangzhou, China
| | - Qi Yu
- Chengdong College, Northeast Agricultural University, Harbin, China
| | - Siqiao Wang
- Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
14
|
Chen H, Luo Y, Li X, Zhang Y, Zheng S, Chen J, Sun Y, Xie Y. The differences of characteristics and allergenicity between natural and recombinant tropomyosin of Macrobrachium nipponense. Food Chem 2024; 460:140610. [PMID: 39068796 DOI: 10.1016/j.foodchem.2024.140610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 07/06/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
Tropomyosin (TM) is the main allergen of Macrobrachium nipponense. Recombinant allergens have great prospects in the detection, diagnosis, and treatment of food allergens. The purpose of this study was to compare the differences in structure and allergenicity between natural TM and recombinant TM. Recombinant TM of M. nipponense with a molecular weight of 38 kDa was successfully expressed in the Escherichia coli system. The amino acid sequence as well as secondary structure between natural and recombinant TM were similar, which were verified by mass and CD spectrometry, respectively. Studies showed that both natural TM and recombinant TM had strong allergenicity, and recombinant TM was more allergenic, which could be used as a substitute for natural TM in the diagnosis and treatment of shrimp allergy. This study provided stable and reliable allergen components for the detection of crustacean allergens and the diagnosis and treatment of food allergies caused by crustacean allergens.
Collapse
Affiliation(s)
- Hongbing Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 Nanjing Dong Road, Nanchang, Jiangxi, 330047, China; Sino German Joint Research Institute, Nanchang University, 235 Nanjing Dong Road, Nanchang, Jiangxi, 330047, China
| | - Yeqing Luo
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 Nanjing Dong Road, Nanchang, Jiangxi, 330047, China; Sino German Joint Research Institute, Nanchang University, 235 Nanjing Dong Road, Nanchang, Jiangxi, 330047, China; College of Food Science and Technology, Nanchang University, 999 Xuefu Road, Nanchang, Jiangxi, 330009, China
| | - Xin Li
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 Nanjing Dong Road, Nanchang, Jiangxi, 330047, China; College of Food Science and Technology, Nanchang University, 999 Xuefu Road, Nanchang, Jiangxi, 330009, China
| | - Yingxue Zhang
- Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI, 48201, USA
| | - Shuangyan Zheng
- Sino German Joint Research Institute, Nanchang University, 235 Nanjing Dong Road, Nanchang, Jiangxi, 330047, China
| | - Jiao Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 Nanjing Dong Road, Nanchang, Jiangxi, 330047, China; Sino German Joint Research Institute, Nanchang University, 235 Nanjing Dong Road, Nanchang, Jiangxi, 330047, China; College of Food Science and Technology, Nanchang University, 999 Xuefu Road, Nanchang, Jiangxi, 330009, China
| | - Yaobin Sun
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 Nanjing Dong Road, Nanchang, Jiangxi, 330047, China; Sino German Joint Research Institute, Nanchang University, 235 Nanjing Dong Road, Nanchang, Jiangxi, 330047, China; College of Food Science and Technology, Nanchang University, 999 Xuefu Road, Nanchang, Jiangxi, 330009, China
| | - Yanhai Xie
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 Nanjing Dong Road, Nanchang, Jiangxi, 330047, China; Sino German Joint Research Institute, Nanchang University, 235 Nanjing Dong Road, Nanchang, Jiangxi, 330047, China.
| |
Collapse
|
15
|
Zhang M, Cui Y, Liu P, Mo R, Wang H, Li Y, Wu Y. Oat β-(1 → 3, 1 → 4)-d-glucan alleviates food allergy-induced colonic injury in mice by increasing Lachnospiraceae abundance and butyrate production. Carbohydr Polym 2024; 344:122535. [PMID: 39218555 DOI: 10.1016/j.carbpol.2024.122535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 07/18/2024] [Accepted: 07/21/2024] [Indexed: 09/04/2024]
Abstract
Oat β-(1 → 3, 1 → 4)-d-glucan (OBG), a linear polysaccharide primarily found in oat bran, has been demonstrated to possess immunomodulatory properties and regulate gut microbiota. This study aimed to investigate the impact of low molecular weight (Mw) OBG (155.2 kDa) on colonic injury and allergic symptoms induced by food allergy (FA), and to explore its potential mechanism. In Experiment 1, results indicated that oral OBG improved colonic inflammation and epithelial barrier, and significantly relieved allergy symptoms. Importantly, the OBG supplement altered the gut microbiota composition, particularly increasing the abundance of Lachnospiraceae and its genera, and promoted the production of short-chain fatty acids, especially butyrate. However, in Experiment 2, the gut microbial depletion eliminated these protective effects of OBG on the colon in allergic mice. Further, in Experiment 3, fecal microbiota transplantation and sterile fecal filtrate transfer directly validated the role of OBG-mediated gut microbiota and its metabolites in relieving FA and its induced colonic injury. Our findings suggest that low Mw OBG can alleviate FA-induced colonic damage by increasing Lachnospiraceae abundance and butyrate production, and provide novel insights into the health benefits and mechanisms of dietary polysaccharide intervention for FA.
Collapse
Affiliation(s)
- Mingrui Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Yingyue Cui
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Pan Liu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Ruixia Mo
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Haotian Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Yingying Li
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Yi Wu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
16
|
Corbett GA, Moore R, Feehily C, Killeen SL, O'Brien E, Van Sinderen D, Matthews E, O'Flaherty R, Rudd PM, Saldova R, Walsh CJ, Lawton EM, MacIntyre DA, Corcoran S, Cotter PD, McAuliffe FM. Dietary amino acids, macronutrients, vaginal birth, and breastfeeding are associated with the vaginal microbiome in early pregnancy. Microbiol Spectr 2024; 12:e0113024. [PMID: 39365058 PMCID: PMC11537119 DOI: 10.1128/spectrum.01130-24] [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: 06/24/2024] [Accepted: 07/29/2024] [Indexed: 10/05/2024] Open
Abstract
The vaginal microbiome is a key player in the etiology of spontaneous preterm birth. This study aimed to illustrate maternal environmental factors associated with vaginal microbiota composition and function in pregnancy. Women in healthy pregnancy had vaginal microbial sampling from the posterior vaginal fornix performed at 16 weeks gestation. After shotgun metagenomic sequencing, heatmaps of relative abundance data were generated. Community state type (CST) was assigned, and alpha diversity was calculated. Demography, obstetric history, well-being, exercise, and diet using food frequency questionnaires were collected and compared against microbial parameters. A total of 119 pregnant participants had vaginal metagenomic sequencing performed. Factors with strongest association with beta diversity were dietary lysine (adj-R2 0.113, P = 0.002), valine (adj-R2 0.096, P = 0.004), leucine (adj-R2 0.086, P = 0.003), and phenylalanine (adj-R2 0.085, P = 0.005, Fig. 2D). Previous vaginal delivery and breastfeeding were associated with vaginal beta diversity (adj-R2 0.048, P = 0.003; adj-R2 0.045, P = 0.004), accounting for 8.5% of taxonomy variation on redundancy analysis. Dietary fat, starch, and maltose were positively correlated with alpha diversity (fat +0.002 SD/g, P = 0.025; starch +0.002 SD/g, P = 0.043; maltose +0.440 SD/g, P = 0.013), particularly in secretor-positive women. Functional signature was associated with CST, maternal smoking, and dietary phenylalanine, accounting for 8.9%-11% of the variation in vaginal microbiome functional signature. Dietary amino acids, previous vaginal delivery, and breastfeeding history were associated with vaginal beta diversity. Functional signature of the vaginal microbiome differed with community state type, smoking, dietary phenylalanine, and vitamin K. Increased alpha diversity correlated with dietary fat and starch. These data provide a novel snapshot into the associations between maternal environment, nutrition, and the vaginal microbiome. IMPORTANCE This secondary analysis of the MicrobeMom randomized controlled trial reveals that dietary amino acids, macronutrients, previous vaginal birth, and breastfeeding have the strongest associations with vaginal taxonomy in early pregnancy. Function of the vaginal niche is associated mainly by species composition, but smoking, vitamin K, and phenylalanine also play a role. These associations provide an intriguing and novel insight into the association between host factors and diet on the vaginal microbiome in pregnancy and highlight the need for further investigation into the complex interactions between the diet, human gut, and vaginal microbiome.
Collapse
Affiliation(s)
- Gillian A. Corbett
- UCD Perinatal Research Centre, UCD School of Medicine, University College Dublin, Dublin, Ireland
- National Maternity Hospital, Dublin 2, Ireland
| | - Rebecca Moore
- UCD Perinatal Research Centre, UCD School of Medicine, University College Dublin, Dublin, Ireland
| | - Conor Feehily
- School of Infection and Immunity, University of Glasgow, Glasgow, United Kingdom
| | - Sarah Louise Killeen
- UCD Perinatal Research Centre, UCD School of Medicine, University College Dublin, Dublin, Ireland
| | - Eileen O'Brien
- School of Biological, Health and Sports Sciences, Technological University Dublin, Dublin, Ireland
| | - Douwe Van Sinderen
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Elizabeth Matthews
- GlycoScience Group, National Institute for Bioprocessing Research and Training (NIBRT), Dublin, Ireland
| | - Roisin O'Flaherty
- GlycoScience Group, National Institute for Bioprocessing Research and Training (NIBRT), Dublin, Ireland
- Department of Chemistry, Maynooth University, Maynooth, Ireland
| | - Pauline M. Rudd
- GlycoScience Group, National Institute for Bioprocessing Research and Training (NIBRT), Dublin, Ireland
- Bioprocessing Technology Institute, AStar, Singapore, Singapore
| | - Radka Saldova
- GlycoScience Group, National Institute for Bioprocessing Research and Training (NIBRT), Dublin, Ireland
- College of Health and Agricultural Science (CHAS), UCD School of Medicine, University College Dublin, Dublin, Ireland
| | - Calum J. Walsh
- Teagasc Food Research Centre, Moorepark, Cork, Ireland
- The Centre for Pathogen Genomics, Department of Microbiology & Immunology, Doherty Institute for Infection & Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | | | - David A. MacIntyre
- Division of the Institute of Reproductive and Developmental Biology, Department of Metabolism, Digestion, and Reproduction, March of Dimes Prematurity Research Centre, Imperial College London, London, United Kingdom
| | - Siobhan Corcoran
- UCD Perinatal Research Centre, UCD School of Medicine, University College Dublin, Dublin, Ireland
- National Maternity Hospital, Dublin 2, Ireland
| | - Paul D. Cotter
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Teagasc Food Research Centre, Moorepark, Cork, Ireland
| | - Fionnuala M. McAuliffe
- UCD Perinatal Research Centre, UCD School of Medicine, University College Dublin, Dublin, Ireland
- National Maternity Hospital, Dublin 2, Ireland
| |
Collapse
|
17
|
Huang J, Wang X, Zhang J, Li Q, Zhang P, Wu C, Jia Y, Su H, Sun X. Fecal microbiota transplantation alleviates food allergy in neonatal mice via the PD-1/PD-L1 pathway and change of the microbiota composition. World Allergy Organ J 2024; 17:100969. [PMID: 39403173 PMCID: PMC11471638 DOI: 10.1016/j.waojou.2024.100969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 08/08/2024] [Accepted: 08/24/2024] [Indexed: 01/03/2025] Open
Abstract
BACKGROUND Food allergy (FA) is a common disorder in children and affects the health of children worldwide. The gut microbiota is closely related to the occurrence and development of FA. Fecal microbiota transplantation (FMT) is a way to treat diseases by reconstituting the microbiota; however, the role and mechanisms of FA have not been validated. METHODS In this study, we established an ovalbumin (OVA)-induced juvenile mouse model and used 16S RNA sequencing, pathological histological staining, molecular biology, and flow-through techniques to evaluate the protective effects of FMT treatment on FA and to explore the mechanisms. RESULTS OVA-induced dysregulation of the gut microbiota led to impaired intestinal function and immune dysregulation in FA mice. FMT treatment improved the structure, diversity, and composition of the gut microbiota and restored it to a near-donor state. FMT treatment reduced levels of Th2-associated inflammatory factors, decreased intestinal tissue inflammation, and reduced IgE production. In addition, FMT reduced the number of mast cells and eosinophils and suppressed OVA-specific antibodies. Further mechanistic studies revealed that FMT treatment induced immune tolerance by inducing the expression of CD103+DCs and programmed cell death ligand 1 (PD-L1) in mesenteric lymph nodes and promoting the production of Treg through the programmed cell death protein 1 (PD-1)/PD-L1 pathway. Meanwhile, Th2 cytokines, OVA-specific antibodies, and PD-1/PD-L1 showed a significant correlation with the gut microbiota. CONCLUSIONS FMT could regulate the gut microbiota and Th1/Th2 immune balance and might inhibit FA through the PD-1/PD-L1 pathway, which would provide a new idea for the treatment of FA.
Collapse
Affiliation(s)
- Jinli Huang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Xingzhi Wang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Juan Zhang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Qiuhong Li
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Panpan Zhang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Cheng Wu
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yuanyuan Jia
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Hui Su
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Xin Sun
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| |
Collapse
|
18
|
Castro AM, Sabater C, Gutiérrez-Díaz I, Navarro S, Rodriguez S, Molinos C, Jiménez S, Claver A, Espin B, Domínguez G, Coronel C, Toyos P, Sariego L, Fernández P, Perez D, Margolles A, Díaz JJ, Delgado S. The intestinal microbiome of infants with cow's milk-induced FPIES is enriched in taxa and genes of enterobacteria. J Pediatr Gastroenterol Nutr 2024; 79:841-849. [PMID: 39175183 DOI: 10.1002/jpn3.12356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/31/2024] [Accepted: 08/07/2024] [Indexed: 08/24/2024]
Abstract
OBJECTIVES Food protein-induced enterocolitis syndrome (FPIES) is a severe type of non-IgE (immunoglobulin E)-mediated (NIM) food allergy, with cow's milk (CM) being the most common offending food. The relationship between the gut microbiota and its metabolites with the inflammatory process in infants with CM FPIES is unknown, although evidence suggests a microbial dysbiosis in NIM patients. This study was performed to contribute to the knowledge of the interaction between the gut microbiota and its derived metabolites with the local immune system in feces of infants with CM FPIES at diagnosis. METHODS Twelve infants with CM FPIES and a matched healthy control group were recruited and the gut microbiota was investigated by 16S amplicon and shotgun sequencing. Fatty acids (FAs) were measured by gas chromatography, while immune factors were determined by enzyme-linked immunosorbent assay and Luminex technology. RESULTS A specific pattern of microbiota in the gut of CM FPIES patients was found, characterized by a high abundance of enterobacteria. Also, an intense excretion of FAs in the feces of these infants was observed. Furthermore, correlations were found between fecal bifidobacteria and immune factors. CONCLUSION These fecal determinations may be useful to gain insight into the pathophysiology of this syndrome and should be taken in consideration for future studies of FPIES patients.
Collapse
Affiliation(s)
- Ana M Castro
- MicroHealth Group, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC)/Instituto Biosanitario del Principado de Asturias (ISPA), Villaviciosa, Asturias, Spain
| | - Carlos Sabater
- MicroHealth Group, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC)/Instituto Biosanitario del Principado de Asturias (ISPA), Villaviciosa, Asturias, Spain
| | - Isabel Gutiérrez-Díaz
- MicroHealth Group, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC)/Instituto Biosanitario del Principado de Asturias (ISPA), Villaviciosa, Asturias, Spain
| | - Sandra Navarro
- Primary Care Center Teatinos-Corredoria, Oviedo, Asturias, Spain
| | - Silvia Rodriguez
- Paediatrics Service, Hospital Universitario de San Agustín, Avilés, Asturias, Spain
| | - Cristina Molinos
- Paediatrics Department, Hospital Universitario de Cabueñes, Gijón, Asturias, Spain
| | | | - Angela Claver
- Allergology, Hospital Universitario Dexeus, Barcelona, Spain
| | - Beatriz Espin
- Paediatric Gastroenterology Unit, Hospital Universitario Virgen del Rocío de Sevilla, Sevilla, Spain
| | - Gloria Domínguez
- Gastroenterology and Nutrition Section, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | | | - Paula Toyos
- Paediatric Group, ISPA, Oviedo, Asturias, Spain
| | - Lydia Sariego
- MicroHealth Group, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC)/Instituto Biosanitario del Principado de Asturias (ISPA), Villaviciosa, Asturias, Spain
| | | | - David Perez
- Paediatrics Service, Hospital Universitario de San Agustín, Avilés, Asturias, Spain
| | - Abelardo Margolles
- MicroHealth Group, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC)/Instituto Biosanitario del Principado de Asturias (ISPA), Villaviciosa, Asturias, Spain
| | - Juan J Díaz
- Paediatric Group, ISPA, Oviedo, Asturias, Spain
| | - Susana Delgado
- MicroHealth Group, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC)/Instituto Biosanitario del Principado de Asturias (ISPA), Villaviciosa, Asturias, Spain
| |
Collapse
|
19
|
Zhou W, Geng Q, Zhang Y, Zhou X, Wu Z, Chen H, El-Sohaimy S. The flavonoid-allergen interaction and its influence on allergenicity. FOOD BIOSCI 2024; 61:104939. [DOI: 10.1016/j.fbio.2024.104939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
20
|
Karimi M, Shirsalimi N, Hashempour Z, Salehi Omran H, Sedighi E, Beigi F, Mortezazadeh M. Safety and efficacy of fecal microbiota transplantation (FMT) as a modern adjuvant therapy in various diseases and disorders: a comprehensive literature review. Front Immunol 2024; 15:1439176. [PMID: 39391303 PMCID: PMC11464302 DOI: 10.3389/fimmu.2024.1439176] [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: 05/28/2024] [Accepted: 09/09/2024] [Indexed: 10/12/2024] Open
Abstract
The human gastrointestinal (GI) tract microbiome is a complex and all-encompassing ecological system of trillions of microorganisms. It plays a vital role in digestion, disease prevention, and overall health. When this delicate balance is disrupted, it can lead to various health issues. Fecal microbiota transplantation (FMT) is an emerging therapeutic intervention used as an adjuvant therapy for many diseases, particularly those with dysbiosis as their underlying cause. Its goal is to restore this balance by transferring fecal material from healthy donors to the recipients. FMT has an impressive reported cure rate between 80% and 90% and has become a favored treatment for many diseases. While FMT may have generally mild to moderate transient adverse effects, rare severe complications underscore the importance of rigorous donor screening and standardized administration. FMT has enormous potential as a practical therapeutic approach; however, additional research is required to further determine its potential for clinical utilization, as well as its safety and efficiency in different patient populations. This comprehensive literature review offers increased confidence in the safety and effectiveness of FMT for several diseases affecting the intestines and other systems, including diabetes, obesity, inflammatory and autoimmune illness, and other conditions.
Collapse
Affiliation(s)
- Mehdi Karimi
- Bogomolets National Medical University (NMU), Kyiv, Ukraine
| | - Niyousha Shirsalimi
- Faculty of Medicine, Hamadan University of Medical Science (UMSHA), Hamadan, Iran
| | - Zahra Hashempour
- School of Medicine, Shiraz University of Medical Sciences (SUMS), Shiraz, Iran
| | - Hossein Salehi Omran
- School of Medicine, Shahid Beheshti University of Medical Sciences (SBMUS), Tehran, Iran
| | - Eshagh Sedighi
- Department of Veterinary Medicine, Islamic Azad University Branch of Urmia, Urmia, Iran
| | - Farzan Beigi
- Students Research Committee, Arak University of Medical Sciences, Arak, Iran
| | - Masoud Mortezazadeh
- Department of Internal Medicine, Sina Hospital, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
21
|
Farnetano M, Carucci L, Coppola S, Oglio F, Masino A, Cozzolino M, Nocerino R, Berni Canani R. Gut microbiome features in pediatric food allergy: a scoping review. FRONTIERS IN ALLERGY 2024; 5:1438252. [PMID: 39386092 PMCID: PMC11461474 DOI: 10.3389/falgy.2024.1438252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 08/13/2024] [Indexed: 10/12/2024] Open
Abstract
Increasing evidence suggests that alterations in the gut microbiome (GM) play a pivotal role in the pathogenesis of pediatric food allergy (FA). This scoping review analyzes the current evidence on GM features associated with pediatric FAs and highlights the importance of the GM as a potential target of intervention for preventing and treating this common condition in the pediatric age. Following the Preferred Reporting Items for Systematic Reviews and Meta-Analysis guidelines, we searched PubMed and Embase using the keywords (gut microbiome OR dysbiosis OR gut microbiota OR microbiome signatures) AND (food allergy OR IgE-mediated food allergy OR food protein-induced allergic proctocolitis OR food protein-induced enterocolitis OR non-IgE food allergy OR cow milk allergy OR hen egg allergy OR peanut allergy OR fish allergy OR shellfish allergy OR tree nut allergy OR soy allergy OR wheat allergy OR rice allergy OR food sensitization). We included 34 studies reporting alterations in the GM in children affected by FA compared with healthy controls. The GM in pediatric FAs is characterized by a higher abundance of harmful microorganisms (e.g., Enterobacteriaceae, Clostridium sensu stricto, Ruminococcus gnavus, and Blautia spp.) and lower abundance of beneficial bacteria (e.g., Bifidobacteriaceae, Lactobacillaceae, some Bacteroides species). Moreover, we provide an overview of the mechanisms of action elicited by these bacterial species in regulating immune tolerance and of the main environmental factors that can modulate the composition and function of the GM in early life. Altogether, these data improve our knowledge of the pathogenesis of FA and can open the way to innovative diagnostic, preventive, and therapeutic strategies for managing these conditions.
Collapse
Affiliation(s)
- Margherita Farnetano
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy
| | - Laura Carucci
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy
- ImmunoNutritionLab at the CEINGE Advanced Biotechnologies Research Center, University of Naples Federico II, Naples, Italy
| | - Serena Coppola
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy
- ImmunoNutritionLab at the CEINGE Advanced Biotechnologies Research Center, University of Naples Federico II, Naples, Italy
| | - Franca Oglio
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy
- ImmunoNutritionLab at the CEINGE Advanced Biotechnologies Research Center, University of Naples Federico II, Naples, Italy
| | - Antonio Masino
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy
- ImmunoNutritionLab at the CEINGE Advanced Biotechnologies Research Center, University of Naples Federico II, Naples, Italy
| | - Marica Cozzolino
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy
- ImmunoNutritionLab at the CEINGE Advanced Biotechnologies Research Center, University of Naples Federico II, Naples, Italy
| | - Rita Nocerino
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy
- ImmunoNutritionLab at the CEINGE Advanced Biotechnologies Research Center, University of Naples Federico II, Naples, Italy
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Rome, Italy
| | - Roberto Berni Canani
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy
- ImmunoNutritionLab at the CEINGE Advanced Biotechnologies Research Center, University of Naples Federico II, Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
- European Laboratory for the Investigation of Food-Induced Diseases, University of Naples Federico II, Naples, Italy
| |
Collapse
|
22
|
Wang Y, Wang B, Sun S, Wang Z. Mapping the relationship between atopic dermatitis and gut microbiota: a bibliometric analysis, 2014-2023. Front Microbiol 2024; 15:1400657. [PMID: 39296293 PMCID: PMC11408322 DOI: 10.3389/fmicb.2024.1400657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 08/26/2024] [Indexed: 09/21/2024] Open
Abstract
Background Atopic dermatitis (AD) is a chronic inflammatory skin condition affecting a significant portion of the population, with prevalence rates of 25% in children and 7-10% in adults. AD not only poses physical challenges but also profoundly impacts patients' mental well-being and quality of life. The stability of gut microbiota is crucial for overall health and can influence AD progression by modulating immune function, skin barrier integrity, and neuroendocrine signaling, which may be an effective target for the prevention and treatment of AD. Thus, exploring the interactions between AD and gut microbiota, particularly in infants, can provide insights into potential preventive and therapeutic strategies. This study aimed to explore the correlation between AD and gut microbiota while providing an overview of current research trends and emerging areas of interest in this field. Methods A comprehensive search was conducted on the Web of Science Core Collection (WOSCC) for relevant publications from January 1, 2014, to December 31, 2023. English-language articles and reviews were included. Two investigators independently screened the publications, and visual analysis was performed using CiteSpace, VOSviewer, Scimago Graphica, and Microsoft Excel software. Results A total of 804 articles were included, showing a significant increase in publications over the past decade. The United States, Wageningen University, and University Ulsan (represented by Hong SJ) had the highest number of published papers. Nutrients was the journal with the most publications, while the Journal of Allergy and Clinical Immunology had the highest number of citations and centrality among co-cited journals. Keyword visualization analysis identified "atopic dermatitis" and "gut microbiota" as central themes. Notably, there has been a notable shift in research focus over the years, with early studies concentrating on "Fecal microbiota," "caesarean section," and "first 6 months," while recent studies have highlighted the roles of "cells," "dysbiosis," and "prebiotics." This shift indicates growing interest in the underlying mechanisms and potential therapeutic interventions related to the intestinal microecology in AD treatment. Conclusion The field of AD and gut microbiota research has evolved significantly, with an increasing focus on understanding the intricate interactions between gut microbiota and AD pathogenesis. Recent years have witnessed increased interest in understanding the relationship between AD and gut microbiota, with researchers conducting extensive studies exploring various aspects of this connection. This review analyzes research trends over the past decade, highlighting trends and hotspots in the study of AD, particularly in infants, and the role of microbiota. This review serves as a valuable reference for future investigations, aiming to provide deeper insights into this burgeoning field and suggests directions for future research.
Collapse
Affiliation(s)
- Yilin Wang
- Department of Dermatology, The 83rd Group Army Hospital of the PLA, Xinxiang, China
| | - Bingkun Wang
- Department of Dermatology, The 83rd Group Army Hospital of the PLA, Xinxiang, China
| | - Shiyou Sun
- Department of Dermatology, The 83rd Group Army Hospital of the PLA, Xinxiang, China
| | - Zhongzhi Wang
- Department of Dermatology, Shanghai Fourth People 's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
23
|
Qiao C, Bian S, Huang H, Xiao H, Ma L, Han R. Impact of ovalbumin allergy on oral and gut microbiome dynamics in 6-week-old BALB/c mice. Front Microbiol 2024; 15:1439452. [PMID: 39290514 PMCID: PMC11406088 DOI: 10.3389/fmicb.2024.1439452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 08/21/2024] [Indexed: 09/19/2024] Open
Abstract
Background The gut microbiota is known to have a significant impact on the development of food allergy, and several recent studies have suggested that both oral microbiota, which first come into contact with allergenic foods, may have a profound influence on the development of food allergy. Methods In this study, we have established an ovalbumin-sensitive mice model by utilizing ovalbumin as a sensitizing agent. Subsequently, we performed a comprehensive analysis of the gut and oral microbiota in ovalbumin-sensitive mice and the control mice using full-length 16S rRNA sequencing analysis. Results Interestingly, both the gut and oral microbiota of ovalbumin-sensitized mice exhibited significant dysbiosis. The relative abundance of s__Lactobacillus_intestinalis in the gut microbiota of ovalbumin-sensitive mice exhibited a significant decrease, whereas the abundance of s__Agrobacterium_radiobacter and s__Acinetobacter_sp__CIP_56_2 displayed a significant increase. Furthermore, the relative abundance of s__unclassified_g__Staphylococcus, s__Streptococcus_hyointestinalis, and s__unclassified_g__Dechloromonas in the oral microbiota of ovalbumin-sensitive mice revealed a significant decrease. In contrast, the abundance of 63 other species, including s__Proteiniclasticum_ruminis, s__Guggenheimella_bovis, and s__Romboutsia_timonensis, demonstrated a significant increase. The random forest classifier achieved the best accuracy in predicting the outcome of food allergy using three gut and three oral biomarkers, with accuracies of 94.12 and 100%, respectively. Based on the predictions of the PICRUSt2 analysis, the only consistent finding observed across multiple samples from both the groups of mice was a significant up-regulation of the nucleotide-binding oligomerization domain (NOD)-like receptor signaling pathway in the ovalbumin-sensitized mice. Conclusion Our study demonstrates that ovalbumin-sensitized mice experience substantial alterations in both gut and oral microbial composition and structure, and specific strains identified in this study may serve as potential biomarkers for food allergy screening. Moreover, our findings highlight that the oral environment, under the same experimental conditions, exhibited greater precision in detecting a larger number of species. Additionally, it is worth noting that the NOD-like receptor signaling pathway plays a vital role in the pathogenesis of OVA (ovalbumin)-induced allergy. These findings will generate novel concepts and strategies in the realm of food allergy prevention and treatment.
Collapse
Affiliation(s)
- Chuanyue Qiao
- Department of Prosthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
- Department of Stomatology, Traditional Chinese Medical Hospital of Huangdao District, Qingdao, China
| | - Shuang Bian
- Department of Prosthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
| | - Hao Huang
- Department of Stomatology, Qingdao Eighth People's Hospital, Qingdao, China
| | - Han Xiao
- Department of Prosthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
| | - Lei Ma
- Department of Prosthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
| | - Rui Han
- Department of Prosthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
| |
Collapse
|
24
|
Ghadimi D, Fölster-Holst R, Blömer S, Ebsen M, Röcken C, Uchiyama J, Matsuzaki S, Bockelmann W. Intricate Crosstalk Between Food Allergens, Phages, Bacteria, and Eukaryotic Host Cells of the Gut-skin Axis. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2024; 97:309-324. [PMID: 39351325 PMCID: PMC11426303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
Bacterial and food allergens are associated with immune-mediated food allergies via the gut-skin axis. However, there has been no data on the potential use of phages to rescue this pathological process. A human triple cell co-culture model incorporating colonocytes (T84 cells), macrophages (THP-1 cells), and hepatocytes (Huh7 cells) was established and infected with Pseudomonas aeruginosa PAO1 (P.a PAO1) in the absence or presence of its KPP22 phage in Dulbecco's Modified Eagle's Medium (DMEM), DMEM+ ovalbumin (OVA), or DMEM+β-casein media. The physiological health of cells was verified by assessing cell viability and Transepithelial electrical resistance (TEER) across the T84 monolayer. The immune response of cells was investigated by determining the secretions of IL-1β, IL-8, IL-22, and IL-25. The ability of P.a PAO1 to adhere to and invade T84 cells was evaluated. The addition of either OVA or β-casein potentiated the P.a PAO1-elicited secretion of cytokines. The viability and TEER of the T84 monolayer were lower in the P.a PAO1+OVA group compared to the P.a PAO1 alone and PAO1+β-casein groups. OVA and β-casein significantly increased the adherence and invasion of P.a PAO1 to T84 cells. In the presence of the KPP22 phage, these disruptive effects were abolished. These results imply that: (1) food allergens and bacterial toxic effector molecules exacerbate each other's disruptive effects; (2) food allergen and bacterial signaling at the gut-skin mucosal surface axis depend on a network of bacteria-phage-eukaryotic host interactions; and (3) phages are complementary for the evaluation of pathobiological processes that occur at the interface between bacteria, host cellular milieu, and food antigens because phages intervene in P.a PAO1-, OVA-, and β-casein-derived inflammation.
Collapse
Affiliation(s)
- Darab Ghadimi
- Department of Microbiology and Biotechnology, Max
Rubner-Institut, Kiel, Germany
| | - Regina Fölster-Holst
- Clinic of Dermatology, Venerology und Allergology,
University Hospital Schleswig-Holstein, Kiel, Germany
| | - Sophia Blömer
- Clinic of Dermatology, Venerology und Allergology,
University Hospital Schleswig-Holstein, Kiel, Germany
| | - Michael Ebsen
- Städtisches MVZ Kiel GmbH (Kiel City Hospital),
Department of Pathology, Kiel, Germany
| | - Christoph Röcken
- Institute of Pathology, Kiel University, University
Hospital, Schleswig-Holstein, Kiel, Germany
| | - Jumpei Uchiyama
- Department of Bacteriology, Graduate School of Medicine
Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Shigenobu Matsuzaki
- Department of Medical Laboratory Science, Faculty of
Health Sciences, Kochi Gakuen University, Kochi, Japan
| | - Wilhelm Bockelmann
- Department of Microbiology and Biotechnology, Max
Rubner-Institut, Kiel, Germany
| |
Collapse
|
25
|
Azzolino D, Verdi L, Perna S, Baldassari I, Cesari M, Lucchi T. Food allergies in older people: An emerging health problem. World Allergy Organ J 2024; 17:100967. [PMID: 39310373 PMCID: PMC11416488 DOI: 10.1016/j.waojou.2024.100967] [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: 09/08/2023] [Revised: 08/14/2024] [Accepted: 08/23/2024] [Indexed: 09/25/2024] Open
Abstract
The prevalence of food allergy (FA) is steadily increasing worldwide. Literature about FA in older people is very scarce since this is predominantly considered as a pediatric condition. However, FA can persist and/or develop across the life course. Given the growing increase in prevalence as well as their persistence, it can be expected that FA will soon represent an important issue in older people. Several age-related factors may potentially mask FA symptoms and/or mediate them in older people. These include mechanisms related to immune senescence, inflammation, and changes in gastrointestinal function as well as micronutrient deficiencies and the use of multiple medications. A multidimensional approach, taking into account the complexity in older people, it is thus important in the evaluation and management of FA during aging. The main and safest strategy in the management of FA is the allergen avoidance since their ingestion may lead to reactions ranging from mild to life-threatening. However, food restrictions should be carefully evaluated, especially in older people, for the risk of nutritional deficiencies and undernutrition.
Collapse
Affiliation(s)
- Domenico Azzolino
- Geriatric Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
| | - Lara Verdi
- Specialization School in Geriatrics, University of Milan, Milan, Italy
| | - Simone Perna
- Department of Food, Environmental and Nutritional Sciences, Division of Human Nutrition, University of Milan, Milan, Italy
| | - Ivan Baldassari
- Department of Research, Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Matteo Cesari
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Tiziano Lucchi
- Geriatric Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
| |
Collapse
|
26
|
Davis EC, Monaco CL, Insel R, Järvinen KM. Gut microbiome in the first 1000 days and risk for childhood food allergy. Ann Allergy Asthma Immunol 2024; 133:252-261. [PMID: 38494114 PMCID: PMC11344696 DOI: 10.1016/j.anai.2024.03.010] [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: 02/19/2024] [Revised: 03/11/2024] [Accepted: 03/11/2024] [Indexed: 03/19/2024]
Abstract
OBJECTIVE To summarize recent data on the association between gut microbiome composition and food allergy (FA) in early childhood and highlight potential host-microbiome interactions that reinforce or abrogate oral tolerance. DATA SOURCES PubMed search of English-language articles related to FA, other atopic disease, and the gut microbiome in pregnancy and early childhood. STUDY SELECTIONS Human studies published after 2015 assessing the relationship between the gut bacteriome and virome in the first 2 years of life and FA or food sensitization development in early childhood were prioritized. Additional human studies conducted on the prenatal gut microbiome or other atopic diseases and preclinical studies are also discussed. RESULTS Children who developed FA harbored lower abundances of Bifidobacterium and Clostridia species and had a less mature microbiome during infancy. The early bacterial microbiome protects against FA through production of anti-inflammatory metabolites and induction of T regulatory cells and may also affect FA risk through a role in trained immunity. Infant enteric phage communities are related to childhood asthma development, though no data are available for FA. Maternal gut microbiome during pregnancy is associated with childhood FA risk, potentially through transplacental delivery of maternal bacterial metabolites, though human studies are lacking. CONCLUSION The maternal and infant microbiomes throughout the first 1000 days of life influence FA risk through a number of proposed mechanisms. Further large, longitudinal cohort studies using taxonomic, functional, and metabolomic analysis of the bacterial and viral microbiomes are needed to provide further insight on the host-microbe interactions underlying FA pathogenesis in childhood.
Collapse
Affiliation(s)
- Erin C Davis
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, New York
| | - Cynthia L Monaco
- Division of Infectious Disease, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York; Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Richard Insel
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, New York
| | - Kirsi M Järvinen
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, New York; Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York; Division of Allergy, Immunology, and Rheumatology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York.
| |
Collapse
|
27
|
Abril AG, Carrera M, Pazos M. Marine Bioactive Compounds with Functional Role in Immunity and Food Allergy. Nutrients 2024; 16:2592. [PMID: 39203729 PMCID: PMC11357426 DOI: 10.3390/nu16162592] [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: 07/16/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 09/03/2024] Open
Abstract
Food allergy, referred to as the atypical physiological overreaction of the immune system after exposure to specific food components, is considered one of the major concerns in food safety. The prevalence of this emerging worldwide problem has been increasing during the last decades, especially in industrialized countries, being estimated to affect 6-8% of young children and about 2-4% of adults. Marine organisms are an important source of bioactive substances with the potential to functionally improve the immune system, reduce food allergy sensitization and development, and even have an anti-allergic action in food allergy. The present investigation aims to be a comprehensive report of marine bioactive compounds with verified actions to improve food allergy and identified mechanisms of actions rather than be an exhaustive compilation of all investigations searching beneficial effects of marine compounds in FA. Particularly, this research highlights the capacity of bioactive components extracted from marine microbial, animal, algae, and microalgae sources, such as n-3 long-chain polyunsaturated fatty acids (LC-PUFA), polysaccharide, oligosaccharide, chondroitin, vitamin D, peptides, pigments, and polyphenols, to regulate the immune system, epigenetic regulation, inflammation, and gut dysbiosis that are essential factors in the sensitization and effector phases of food allergy. In conclusion, the marine ecosystem is an excellent source to provide foods with the capacity to improve the hypersensitivity induced against specific food allergens and also bioactive compounds with a potential pharmacological aptitude to be applied as anti-allergenic in food allergy.
Collapse
Affiliation(s)
- Ana G. Abril
- Department of Microbiology and Parasitology, Faculty of Pharmacy, University of Santiago de Compostela, 15898 Santiago de Compostela, Spain;
- Institute of Marine Research (IIM-CSIC), Spanish National Research Council (CSIC), 36208 Vigo, Spain;
| | - Mónica Carrera
- Institute of Marine Research (IIM-CSIC), Spanish National Research Council (CSIC), 36208 Vigo, Spain;
| | - Manuel Pazos
- Institute of Marine Research (IIM-CSIC), Spanish National Research Council (CSIC), 36208 Vigo, Spain;
| |
Collapse
|
28
|
Ponda P, Cerise JE, Navetta-Modrov B, Kiehm J, Covelli GM, Weiss J, Lee AT. The age-specific microbiome of children with milk, egg, and peanut allergy. Ann Allergy Asthma Immunol 2024; 133:203-210.e6. [PMID: 38697287 DOI: 10.1016/j.anai.2024.04.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/10/2024] [Accepted: 04/22/2024] [Indexed: 05/04/2024]
Abstract
BACKGROUND Immune regulation by gut microbiota is affected by dysbiosis and may precede food allergy onset. Prior studies lacked comparisons stratified by age and clinical phenotype. OBJECTIVE To assess the microbiome of children with food allergy (<3 years, 3-18 years) compared with similar aged children without food allergy. METHODS A real-world prospective cross-sectional study performed from 2014 to 2019 recruited children highly likely to have milk, egg, or peanut allergy defined by history and serum IgE or confirmed by food challenge. 16S ribosomal RNA sequencing identified stool microbial DNA. Alpha and beta diversity was compared between groups with food allergy and healthy controls stratified by age. Differential abundance for non a priori taxa was accepted at absolute fold-change greater than 2 and q value less than 0.05. RESULTS A total of 70 patients were included (56 with food allergy and 14 healthy controls). Groups were not significantly different in age, gender at birth, race, mode of delivery, breastfeeding duration, or antibiotic exposure. Younger children with food allergy had similar alpha diversity compared with controls. Beta diversity was significantly different by age (P = .001). There was differential abundance of several a priori (P < .05) taxa (including Clostridia) only in younger children. Both a priori (including Coprococcus and Clostridia) and non a priori (q < 0.05) Acidobacteria_Gp15, Aestuariispira, Tindallia, and Desulfitispora were significant in older children with food allergy, especially with peanut allergy. CONCLUSION Dysbiosis associates with food allergy, most prominent in older children with peanut allergy. Younger children with and without food allergy have fewer differences in gut microbiota. This correlates with clinical observations of persistence of peanut allergy and improved efficacy and safety of oral immunotherapy in younger children. Age younger than 3 years should be considered when initiating therapeutic interventions.
Collapse
Affiliation(s)
- Punita Ponda
- Northwell, New Hyde Park, New York; Division of Allergy and Immunology, Cohen Children's Medical Center, New Hyde Park, New York.
| | - Jane E Cerise
- Biostatistics Unit, Office of Academic Affairs, Northwell Health, New Hyde Park, New York
| | - Brianne Navetta-Modrov
- Department of Medicine, Stony Brook University School of Medicine, Stony Brook, New York
| | - Jamie Kiehm
- Los Angeles County Department of Health Services, Los Angeles, California
| | - Grace M Covelli
- Walter Reed National Military Medical Center, Bethesda, Maryland
| | - Jared Weiss
- Department of Psychiatry, New York University School of Medicine, New York, New York
| | - Annette T Lee
- Northwell, New Hyde Park, New York; Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York
| |
Collapse
|
29
|
Peer A, Samuelson DR. The Role of the Microbiome in Allergy, Asthma, and Occupational Lung Disease. Curr Allergy Asthma Rep 2024; 24:415-423. [PMID: 38904934 PMCID: PMC11297072 DOI: 10.1007/s11882-024-01156-8] [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] [Accepted: 06/07/2024] [Indexed: 06/22/2024]
Abstract
PURPOSE OF REVIEW The human commensal microbiota is now widely accepted as a key regulator of human health and disease. The composition of the mucosal associated microbiota has been shown to play a critical role in the lung health. The role of the mucosal microbiota in the development and severity of allergy, asthma, and occupational lung disease is only beginning to take shape. However, advances in our understanding of these links have tremendous potential to led to new clinical interventions to reduce allergy, asthma, and occupational lung disease morbidity. RECENT FINDINGS We review recent work describing the relationship and role of the commensal microbiota in the development of allergy, asthma, and occupational lung disease. Our review primarily focuses on occupational exposures and the effects of the microbiome, both in composition and function. Data generated from these studies may lead to the development of interventions targeted at establishing and maintaining a healthy microbiota. We also highlight the role of environmental exposures and the effects on the commensal microbial community and their potential association with occupational lung disease. This review explores the current research describing the role of the human microbiome in the regulation of pulmonary health and disease, with a specific focus on the role of the mucosal microbiota in the development of allergy, asthma, and occupational lung disease.
Collapse
Affiliation(s)
- Ashley Peer
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep, University of Nebraska Medical Center, Omaha, NE, USA
| | - Derrick R Samuelson
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep, University of Nebraska Medical Center, Omaha, NE, USA.
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, NE, USA.
| |
Collapse
|
30
|
Zheng W, Wai CYY, Sit JKC, Cheng NS, Leung CWM, Leung TF. Routinely Used and Emerging Diagnostic and Immunotherapeutic Approaches for Wheat Allergy. Biomedicines 2024; 12:1549. [PMID: 39062122 PMCID: PMC11275021 DOI: 10.3390/biomedicines12071549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 07/06/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Wheat, a component of the staple diet globally, is a common food allergen in children. The symptoms of wheat allergy (WA) range from skin rash to shortness of breath, significantly impairing quality of life. Following initial clinical suspicion, individuals may undergo routinely used allergy tests such as a wheat allergen-specific skin prick test (SPT), a blood test for specific immunoglobulin E (sIgE) levels, or oral food challenge. Conventional management of WA lies in wheat avoidance, yet accidental consumption may be inevitable owing to the ubiquity of wheat in various food products. This article aims to provide an overview of the immunologic pathway of WA, followed by its emerging diagnostic methods, namely alcohol-soluble SPT extracts, component-resolved diagnosis, and the basophil activation test (BAT). The mechanisms underlying wheat allergen-specific oral immunotherapy (OIT) as well as a summary of the efficacy, tolerability, and safety of related clinical trials will then be discussed.
Collapse
Affiliation(s)
- Wanqi Zheng
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong; (W.Z.); (C.Y.Y.W.); (J.K.C.S.); (N.S.C.)
| | - Christine Yee Yan Wai
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong; (W.Z.); (C.Y.Y.W.); (J.K.C.S.); (N.S.C.)
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Jason Ka Chun Sit
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong; (W.Z.); (C.Y.Y.W.); (J.K.C.S.); (N.S.C.)
| | - Nam Sze Cheng
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong; (W.Z.); (C.Y.Y.W.); (J.K.C.S.); (N.S.C.)
| | | | - Ting Fan Leung
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong; (W.Z.); (C.Y.Y.W.); (J.K.C.S.); (N.S.C.)
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
31
|
Murdaca G, Tagliafico L, Page E, Paladin F, Gangemi S. Gender Differences in the Interplay between Vitamin D and Microbiota in Allergic and Autoimmune Diseases. Biomedicines 2024; 12:1023. [PMID: 38790985 PMCID: PMC11117902 DOI: 10.3390/biomedicines12051023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/22/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024] Open
Abstract
The synergic role of vitamin D and the intestinal microbiota in the regulation of the immune system has been thoroughly described in the literature. Vitamin D deficiency and intestinal dysbiosis have shown a pathogenetic role in the development of numerous immune-mediated and allergic diseases. The physiological processes underlying aging and sex have proven to be capable of having a negative influence both on vitamin D values and the biodiversity of the microbiome. This leads to a global increase in levels of systemic inflammatory markers, with potential implications for all immune-mediated diseases and allergic conditions. Our review aims to collect and analyze the relationship between vitamin D and the intestinal microbiome with the immune system and the diseases associated with it, emphasizing the effect mediated by sexual hormones and aging.
Collapse
Affiliation(s)
- Giuseppe Murdaca
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy (E.P.)
- Allergology and Clinical Immunology Unit, San Bartolomeo Hospital, 19038 Sarzana, Italy
| | - Luca Tagliafico
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy (E.P.)
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Elena Page
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy (E.P.)
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Francesca Paladin
- Elderly and Disabeld Department, San Paolo Hospital, 17100 Savona, Italy
| | - Sebastiano Gangemi
- School and Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| |
Collapse
|
32
|
Murali SK, Mansell TJ. Next generation probiotics: Engineering live biotherapeutics. Biotechnol Adv 2024; 72:108336. [PMID: 38432422 DOI: 10.1016/j.biotechadv.2024.108336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 02/10/2024] [Accepted: 02/25/2024] [Indexed: 03/05/2024]
Abstract
The population dynamics of the human microbiome have been associated with inflammatory bowel disease, cancer, obesity, autoimmune diseases, and many other human disease states. An emerging paradigm in treatment is the administration of live engineered organisms, also called next-generation probiotics. However, the efficacy of these microbial therapies can be limited by the organism's overall performance in the harsh and nutrient-limited environment of the gut. In this review, we summarize the current state of the art use of bacterial and yeast strains as probiotics, highlight the recent development of genetic tools for engineering new therapeutic functions in these organisms, and report on the latest therapeutic applications of engineered probiotics, including recent clinical trials. We also discuss the supplementation of prebiotics as a method of manipulating the microbiome and improving the overall performance of engineered live biotherapeutics.
Collapse
Affiliation(s)
- Sanjeeva Kumar Murali
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, USA.
| | - Thomas J Mansell
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, USA; Interdepartmental Microbiology Graduate Program, Iowa State University, Ames, IA 50011, USA.
| |
Collapse
|
33
|
Kuźniar J, Kozubek P, Gomułka K. Differences in the Course, Diagnosis, and Treatment of Food Allergies Depending on Age-Comparison of Children and Adults. Nutrients 2024; 16:1317. [PMID: 38732564 PMCID: PMC11085589 DOI: 10.3390/nu16091317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/23/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
Food allergy (FA) has become a common global public health issue, with a growing prevalence in the modern world and a significant impact on the lives of patients, their families, and caregivers. It affects every area of life and is associated with elevated costs. Food allergy is an adverse immune reaction that occurs in response to a given food. The symptoms vary from mild to severe and can lead to anaphylaxis. This is why it is important to focus on the factors influencing the occurrence of food allergies, specific diagnostic methods, effective therapies, and especially prevention. Recently, many guidelines have emphasized the impact of introducing specific foods into a child's diet at an early age in order to prevent food allergies. Childhood allergies vary with age. In infants, the most common allergy is to cow's milk. Later in life, peanut allergy is more frequently diagnosed. Numerous common childhood allergies can be outgrown by adulthood. Adults can also develop new IgE-mediated FA. The gold standard for diagnosis is the oral provocation test. Skin prick tests, specific IgE measurements, and component-resolved diagnostic techniques are helpful in the diagnosis. Multiple different approaches are being tried as possible treatments, such as immunotherapy or monoclonal antibodies. This article focuses on the prevention and quality of life of allergic patients. This article aims to systematize the latest knowledge and highlight the differences between food allergies in pediatric and adult populations.
Collapse
Affiliation(s)
- Julia Kuźniar
- Student Scientific Group of Internal Medicine and Allergology, Wroclaw Medical University, 50-369 Wroclaw, Poland;
| | - Patrycja Kozubek
- Student Scientific Group of Internal Medicine and Allergology, Wroclaw Medical University, 50-369 Wroclaw, Poland;
| | - Krzysztof Gomułka
- Department of Internal Medicine, Pneumology and Allergology, Wroclaw Medical University, 50-369 Wroclaw, Poland;
| |
Collapse
|
34
|
Castro AM, Gutiérrez-Díaz I, Saiz ML, Navarro S, Suárez M, Carbajal I, García Á, Sariego L, Toyos P, Rodríguez S, Jiménez S, González D, Molinos C, Pérez D, Fernández P, Suárez-Alvarez B, Margolles A, Díaz JJ, Delgado S. Gut microbiota and inflammatory mediators differentiate IgE mediated and non-IgE mediated cases of cow's milk protein at diagnosis. J Pediatr Gastroenterol Nutr 2024; 78:836-845. [PMID: 38344848 DOI: 10.1002/jpn3.12155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/12/2024] [Accepted: 01/24/2024] [Indexed: 03/08/2024]
Abstract
OBJECTIVE Analyze fecal and blood samples at point of diagnosis in IgE mediated cow's milk protein allergy (CMPA) and non-IgE mediated (NIM)-CMPA patients to look for potential new biomarkers. PATIENTS AND METHODS Fourteen patients with IgE mediated CMPA and 13 with NIM-CMPA were recruited in three hospitals in the north of Spain, and were compared with 25 infants from a control group of the same age range. To characterize intestinal microbiota, 16S rDNA gene and internal transcribed spacer amplicons of bifidobacteria were sequenced with Illumina technology. Fatty acids were analyzed by gas chromatography, meanwhile intestinal inflammation markers were quantified by enzyme-linked immunosorbent assay and a multiplex system. Immunological analysis of blood was performed by flow cytometry. RESULTS The fecal results obtained in the NIM-CMPA group stand out. Among them, a significant reduction in the abundance of Bifidobacteriaceae and Bifidobacterium sequences with respect to controls was observed. Bifidobacterial species were also different, highlighting the lower abundance of Bifidobacterium breve sequences. Fecal calprotectin levels were found to be significantly elevated in relation to IgE mediated patients. Also, a higher excretion of IL-10 and a lower excretion of IL-1ra and platelet derived growth factor-BB was found in NIM-CMPA patients. CONCLUSIONS The differential fecal parameters found in NIM-CMPA patients could be useful in the diagnosis of NIM food allergy to CM proteins.
Collapse
Affiliation(s)
- Ana M Castro
- MicroHealth Group, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC)/Instituto Biosanitario del Principado de Asturias (ISPA), Villaviciosa, Asturias, Spain
| | - Isabel Gutiérrez-Díaz
- MicroHealth Group, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC)/Instituto Biosanitario del Principado de Asturias (ISPA), Villaviciosa, Asturias, Spain
| | - María L Saiz
- Translational Immunology, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Oviedo, Asturias, Spain
| | - Sandra Navarro
- Primary Care Center Teatinos-Corredoria, Oviedo, Asturias, Spain
| | | | | | - Águeda García
- Primary Care Center Vallobin-La Florida, Oviedo, Asturias, Spain
| | - Lydia Sariego
- MicroHealth Group, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC)/Instituto Biosanitario del Principado de Asturias (ISPA), Villaviciosa, Asturias, Spain
| | - Paula Toyos
- Pediatric Group, ISPA, Oviedo, Asturias, Spain
| | - Silvia Rodríguez
- Pediatrics Service, Hospital Universitario de San Agustín, Avilés, Asturias, Spain
| | | | | | - Cristina Molinos
- Pediatrics Department, Hospital Universitario de Cabueñes, Gijón, Asturias, Spain
| | - David Pérez
- Pediatrics Service, Hospital Universitario de San Agustín, Avilés, Asturias, Spain
| | | | - Beatriz Suárez-Alvarez
- Translational Immunology, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Oviedo, Asturias, Spain
| | - Abelardo Margolles
- MicroHealth Group, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC)/Instituto Biosanitario del Principado de Asturias (ISPA), Villaviciosa, Asturias, Spain
| | - Juan J Díaz
- Pediatric Group, ISPA, Oviedo, Asturias, Spain
| | - Susana Delgado
- MicroHealth Group, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC)/Instituto Biosanitario del Principado de Asturias (ISPA), Villaviciosa, Asturias, Spain
| |
Collapse
|
35
|
Cho H, Kim J, Kim S, Jeong HI, Kwon M, Kim HM, Shim JS, Kim K, Baek J, Kyung Y, Choi SJ, Oh SY, Bae J, Won HH, Kim J, Ahn K. Postpartum Maternal Anxiety Affects the Development of Food Allergy Through Dietary and Gut Microbial Diversity During Early Infancy. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2024; 16:154-167. [PMID: 38528383 DOI: 10.4168/aair.2024.16.2.154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 11/29/2023] [Accepted: 12/25/2023] [Indexed: 03/27/2024]
Abstract
PURPOSE We aimed to investigate the mediating factors between maternal anxiety and the development of food allergy (FA) in children until 2 years from birth. METHODS In this longitudinal cohort of 122 mother-child dyads from pregnancy to 24 months of age, we regularly surveyed maternal psychological states, infant feeding data, and allergic symptoms and collected stool samples at 6 months of age for microbiome analysis. Considering the temporal order of data collection, we investigated serial mediating effects and indirect effects among maternal anxiety, dietary diversity (DD), gut microbial diversity, and FA using structural equation modeling. RESULTS Among the 122 infants, 15 (12.3%) were diagnosed with FA. Increased maternal anxiety between 3 and 6 months after delivery was associated with a lower DD score. Infants with low DD at 4 months showed low gut microbial richness, which was associated with FA development. When the infants were grouped into 4 subtypes, using consensus clustering of 13 gut bacteria significantly associated with maternal anxiety and DD, Prevotella, Eubacterium, Clostridiales and Lachnospiraceae were more abundant in the group with lower FA occurrence. CONCLUSIONS Postpartum maternal anxiety, mediated by reduced DD and gut microbial diversity, may be a risk factor for the development of FA in infants during the first 2 years of life.
Collapse
Affiliation(s)
- Hyunbin Cho
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea
| | - Jiwon Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sukyung Kim
- Department of Pediatrics, Hallym University Dongtan Sacred Heart Hospital, Hallym University School of Medicine, Hwaseong, Korea
| | - Hye-In Jeong
- Department of Pediatrics, Eulji University Hospital, Eulji University School of Medicine, Seoul, Korea
| | - Mijeong Kwon
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hyun Mi Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ji Sun Shim
- Statistics and Data Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Kyunga Kim
- Statistics and Data Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Jihyun Baek
- Department of Psychology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yechan Kyung
- Department of Pediatrics, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Korea
| | - Suk-Joo Choi
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Soo-Young Oh
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jaewoong Bae
- R&D Institute, BioEleven Co., Ltd., Seoul, Korea
| | - Hong-Hee Won
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea.
| | - Jihyun Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Seoul, Korea.
| | - Kangmo Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Seoul, Korea
| |
Collapse
|
36
|
Duan C, Ma L, Qin M, Zhang L, Hu S, Liu L, Sun Y, Ma F, Li D. Potential of Lactobacillus plantarum A56 in relieving food allergy through immunoregulation, antioxidation, and reshaping intestinal microbiota. J Nutr Biochem 2024; 125:109560. [PMID: 38163625 DOI: 10.1016/j.jnutbio.2023.109560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 11/14/2023] [Accepted: 12/28/2023] [Indexed: 01/03/2024]
Abstract
Food allergy is an abnormal immune reaction triggered by food protein antigens. Relevant studies have suggested that probiotic supplementation was with the potential to alleviate food allergy. This study aimed to explore the effects of Lactobacillus plantarum A56 on the alleviation of ovalbumin (OVA)-induced food allergy via immunomodulatory function, antioxidation, and modification of intestinal microbiota. Balb/c mice were sensitized with OVA (20 µg/mouse) by intraperitoneal injection for 3 weeks and accompanied by oral administration of L. plantarum A56 (109 CFU/mL), subsequently with orally challenged twice by OVA at 50 mg/mL for 1 week. The results showed that oral supplementation of L. plantarum A56 could effectively relieve allergic symptoms of mice, and decreased OVA-specific IgE and IgG1 concentrations. It also declined interleukin (IL)-4 level, raised interferon-γ (IFN-γ) in serum, and splenocyte supernatant, and the qPCR results were consistent with above results. Moreover, L. plantarum A56 treatment also fortified superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) levels, and reduced malondialdehyde (MDA) level in serum. The increased nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and forkhead box O1 (Foxo1) expression indicated that L. plantarum A56 exerted antioxidation through Nrf2-Foxo1 pathway. In addition, L. plantarum A56 treatment elevated Bacteroidetes richness, ASV/OTU number, species diversity, etc. Notably, Spearman correlation analysis indicated that Bacteroidetes displayed obviously negative correlation with IgE and IgG1, but Actinobacteria and Acidobacteria exhibited significantly positive correlation with IgG1 and IgE. Collectively, these results suggested that L. plantarum A56 could alleviate OVA-induced food allergy by regulating Th1/Th2 imbalance, antioxidation, and modulating intestinal microbiota.
Collapse
Affiliation(s)
- Cuicui Duan
- Key Laboratory of Agro-products Processing Technology, Jilin Provincial Department of Education, Changchun University, Changchun, Jilin, People's Republic of China; Key Laboratory of Intelligent Rehabilitation and Barrier-free for the Disabled, Ministry of Education, Changchun University, Changchun, Jilin, People's Republic of China
| | - Lin Ma
- Key Laboratory of Agro-products Processing Technology, Jilin Provincial Department of Education, Changchun University, Changchun, Jilin, People's Republic of China
| | - Mengchun Qin
- Key Laboratory of Agro-products Processing Technology, Jilin Provincial Department of Education, Changchun University, Changchun, Jilin, People's Republic of China
| | - Lingfang Zhang
- Key Laboratory of Agro-products Processing Technology, Jilin Provincial Department of Education, Changchun University, Changchun, Jilin, People's Republic of China
| | - Shunan Hu
- Key Laboratory of Agro-products Processing Technology, Jilin Provincial Department of Education, Changchun University, Changchun, Jilin, People's Republic of China
| | - Lifan Liu
- Key Laboratory of Agro-products Processing Technology, Jilin Provincial Department of Education, Changchun University, Changchun, Jilin, People's Republic of China
| | - Yixue Sun
- Key Laboratory of Agro-products Processing Technology, Jilin Provincial Department of Education, Changchun University, Changchun, Jilin, People's Republic of China
| | - Fumin Ma
- Key Laboratory of Agro-products Processing Technology, Jilin Provincial Department of Education, Changchun University, Changchun, Jilin, People's Republic of China
| | - Dan Li
- Key Laboratory of Agro-products Processing Technology, Jilin Provincial Department of Education, Changchun University, Changchun, Jilin, People's Republic of China; Key Laboratory of Intelligent Rehabilitation and Barrier-free for the Disabled, Ministry of Education, Changchun University, Changchun, Jilin, People's Republic of China.
| |
Collapse
|
37
|
Ataya J, Ismail A, Soqia J, Kousa A, Shahoud Z, Alhalabi R, Zaitouna M. Addressing knowledge gaps in allergies among Syrian hospital patients: a cross-sectional study. Sci Rep 2024; 14:2938. [PMID: 38316903 PMCID: PMC10844623 DOI: 10.1038/s41598-024-53471-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 01/31/2024] [Indexed: 02/07/2024] Open
Abstract
Allergies have a significant impact on health and quality of life worldwide, yet there is limited research on the awareness and knowledge of allergies. This study aimed to explore the level of awareness and knowledge of allergies among visitors in Syrian hospitals. A cross-sectional study was conducted between May and September 2022, and a standardized international questionnaire was administered to 504 visitors in three hospitals in Syria. Data analysis was performed using the Statistical Package for Social Sciences (SPSS Inc., Chicago, IL, USA) version 23. The final sample comprised 504 questionnaires with 61.7% of participants achieving an average score. Statistical analysis revealed a significant difference in knowledge of allergy scores between the group with only elementary education (M = 3.76, SD = 1.67, p = 0.011), the group with no education (M = 3.65, SD = 1.65, p = 0.006), and the group with a university education (M = 4.44, SD = 1.25). Notably, no significant differences were found between the other educational groups. A one-way ANOVA was employed to assess the effect of place of living on knowledge of allergy, but no significant differences were observed between the groups (p = 0.462). Lastly, a significant negative correlation was detected between participant age and knowledge of allergy scores (r(502) = - 0.102, p = 0.022). Allergies represent a substantial global health concern that demands attention from healthcare providers, policymakers, and the public. This study emphasizes the importance of investing in health education and awareness campaigns to enhance knowledge and comprehension of allergies, particularly individuals with lower levels of education levels. By equipping individuals with the requisite information to effectively manage their allergies, their overall health and wellbeing can be improved.
Collapse
Affiliation(s)
- Jamal Ataya
- Faculty of Medicine, University of Aleppo, Aleppo, Syria.
| | | | - Jameel Soqia
- Faculty of Medicine, Damascus University, Damascus, Syria
| | - Alyamama Kousa
- Faculty of Medicine, Damascus University, Damascus, Syria
| | - Zein Shahoud
- Faculty of Medicine, Damascus University, Damascus, Syria
| | - Rawan Alhalabi
- Faculty of Medicine, Damascus University, Damascus, Syria
| | - Mazen Zaitouna
- Faculty of Medicine, Damascus University, Damascus, Syria
- UMR1195, University Paris Sud, INSERM, University Paris-Saclay, Le Kremlin-Bicetre, France
- Service of Urology, Grand hospital of Est Francilian, Meaux, France
| |
Collapse
|
38
|
Saroukhani S, Samms-Vaughan M, Bressler J, Lee M, Byrd-Williams C, Hessabi M, Grove ML, Shakespeare-Pellington S, Loveland KA, Rahbar MH. Additive or Interactive Associations of Food Allergies with Glutathione S-Transferase Genes in Relation to ASD and ASD Severity in Jamaican Children. J Autism Dev Disord 2024; 54:704-724. [PMID: 36436147 DOI: 10.1007/s10803-022-05813-7] [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] [Accepted: 10/30/2022] [Indexed: 11/29/2022]
Abstract
To investigate additive and interactive associations of food allergies with three glutathione S-transferase (GST) genes in relation to ASD and ASD severity in Jamaican children. Using data from 344 1:1 age- and sex-matched ASD cases and typically developing controls, we assessed additive and interactive associations of food allergies with polymorphisms in GST genes (GSTM1, GSTP1 and GSTT1) in relation to ASD by applying conditional logistic regression models, and in relation to ASD severity in ASD cases as measured by the Autism Diagnostic Observation Schedule-2nd Edition (ADOS-2) total and domains specific comparison scores (CSs) by fitting general linear models. Although food allergies and GST genes were not associated with ASD, ASD cases allergic to non-dairy food had higher mean ADOS-2 Restricted and Repetitive Behaviors (RRB) CS (8.8 vs. 8.0, P = 0.04). In addition, allergy to dairy was associated with higher mean RRB CS only among ASD cases with GSTT1 DD genotype (9.9 vs. 7.8, P < 0.01, interaction P = 0.01), and GSTP1 Val/Val genotype under a recessive genetic model (9.8 vs. 7.8, P = 0.02, interaction P = 0.06). Our findings are consistent with the role for GST genes in ASD and food allergies, though require replication in other populations.
Collapse
Affiliation(s)
- Sepideh Saroukhani
- Division of Clinical and Translational Sciences, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
- Biostatistics/Epidemiology/Research Design (BERD) Core, Center for Clinical and Translational Sciences (CCTS), The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Maureen Samms-Vaughan
- Department of Child & Adolescent Health, The University of the West Indies (UWI), Mona Campus, Kingston 7, Kingston, Jamaica
| | - Jan Bressler
- Department of Epidemiology, Human Genetics, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - MinJae Lee
- Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Courtney Byrd-Williams
- Department of Health Promotion and Behavioral Sciences, Michael & Susan Dell Center for Healthy Living, School of Public Health Regional Campus at Austin, The University of Texas Health Science Center at Houston, Austin, TX, 78701, USA
| | - Manouchehr Hessabi
- Biostatistics/Epidemiology/Research Design (BERD) Core, Center for Clinical and Translational Sciences (CCTS), The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Megan L Grove
- Department of Epidemiology, Human Genetics, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Sydonnie Shakespeare-Pellington
- Department of Child & Adolescent Health, The University of the West Indies (UWI), Mona Campus, Kingston 7, Kingston, Jamaica
| | - Katherine A Loveland
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, TX, 77030, Houston, USA
| | - Mohammad H Rahbar
- Division of Clinical and Translational Sciences, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
- Biostatistics/Epidemiology/Research Design (BERD) Core, Center for Clinical and Translational Sciences (CCTS), The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
- Department of Epidemiology, Human Genetics, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| |
Collapse
|
39
|
Savova MV, Zhu P, Harms AC, van der Molen RG, Belzer C, Hendrickx DM. Current insights into cow's milk allergy in children: Microbiome, metabolome, and immune response-A systematic review. Pediatr Allergy Immunol 2024; 35:e14084. [PMID: 38363041 DOI: 10.1111/pai.14084] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/17/2024]
Abstract
The increasing prevalence of IgE-mediated cow's milk allergy (CMA) in childhood is a worldwide health concern. There is a growing awareness that the gut microbiome (GM) might play an important role in CMA development. Therefore, treatment with probiotics and prebiotics has gained popularity. This systematic review provides an overview of the alterations of the GM, metabolome, and immune response in CMA children and animal models, including post-treatment modifications. MEDLINE, PubMed, Scopus, and Web of Science were searched for studies on GM in CMA-diagnosed children, published before 1 March 2023. A total of 21 articles (13 on children and 8 on animal models) were included. The studies suggest that the GM, characterized by an enrichment of the Clostridia class and reductions in the Lactobacillales order and Bifidobacterium genus, is associated with CMA in early life. Additionally, reduced levels of short-chain fatty acids (SCFAs) and altered amino acid metabolism were reported in CMA children. Commonly used probiotic strains belong to the Bifidobacterium and Lactobacillus genera. However, only Bifidobacterium levels were consistently upregulated after the intervention, while alterations of other bacteria taxa remain inconclusive. These interventions appear to contribute to the restoration of SCFAs and amino acid metabolism balance. Mouse models indicate that these interventions tend to restore the Th 2/Th 1 balance, increase the Treg response, and/or silence the overall pro- and anti-inflammatory cytokine response. Overall, this systematic review highlights the need for multi-omics-related research in CMA children to gain a mechanistic understanding of this disease and to develop effective treatments and preventive strategies.
Collapse
Affiliation(s)
- Mariyana V Savova
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Pingping Zhu
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Amy C Harms
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Renate G van der Molen
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboudumc, Nijmegen, The Netherlands
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Diana M Hendrickx
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| |
Collapse
|
40
|
Cao S, Maulloo CD, Raczy MM, Sabados M, Slezak AJ, Nguyen M, Solanki A, Wallace RP, Shim HN, Wilson DS, Hubbell JA. Glycosylation-modified antigens as a tolerance-inducing vaccine platform prevent anaphylaxis in a pre-clinical model of food allergy. Cell Rep Med 2024; 5:101346. [PMID: 38128531 PMCID: PMC10829738 DOI: 10.1016/j.xcrm.2023.101346] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 09/06/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023]
Abstract
The only FDA-approved oral immunotherapy for a food allergy provides protection against accidental exposure to peanuts. However, this therapy often causes discomfort or side effects and requires long-term commitment. Better preventive and therapeutic solutions are urgently needed. We develop a tolerance-inducing vaccine technology that utilizes glycosylation-modified antigens to induce antigen-specific non-responsiveness. The glycosylation-modified antigens are administered intravenously (i.v.) or subcutaneously (s.c.) and traffic to the liver or lymph nodes, respectively, leading to preferential internalization by antigen-presenting cells, educating the immune system to respond in an innocuous way. In a mouse model of cow's milk allergy, treatment with glycosylation-modified β-lactoglobulin (BLG) is effective in preventing the onset of allergy. In addition, s.c. administration of glycosylation-modified BLG shows superior safety and potential in treating existing allergies in combination with anti-CD20 co-therapy. This platform provides an antigen-specific immunomodulatory strategy to prevent and treat food allergies.
Collapse
Affiliation(s)
- Shijie Cao
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA; Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA.
| | - Chitavi D Maulloo
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Michal M Raczy
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Matthew Sabados
- Biological Sciences Division, University of Chicago, Chicago, IL 60637, USA
| | - Anna J Slezak
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Mindy Nguyen
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA; Animal Resource Center, University of Chicago, Chicago, IL 60637, USA
| | - Ani Solanki
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA; Animal Resource Center, University of Chicago, Chicago, IL 60637, USA
| | - Rachel P Wallace
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Ha-Na Shim
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - D Scott Wilson
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA; Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA
| | - Jeffrey A Hubbell
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA; Committee on Immunology, University of Chicago, Chicago, IL 60637, USA; Committee on Cancer Biology, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
41
|
Hazan G, Goldstein Y, Greenberg D, Khalde F, Mahajna R, Keren-Naos A, Hershkovitz E, Faingelernt Y, Givon-Lavi N, Danino D. Comparing single versus multiple virus detection in pediatric acute gastroenteritis postimplementation of routine multiplex RT-PCR diagnostic testing. J Med Virol 2024; 96:e29344. [PMID: 38149453 DOI: 10.1002/jmv.29344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/27/2023] [Accepted: 12/13/2023] [Indexed: 12/28/2023]
Abstract
Utilizing multiplex real time polymerase chain reaction (RT-PCR) for rapid diagnosis of gastroenteritis, enables simultaneous detection of multiple pathogens. A comparative analysis of disease characteristics was conducted between cases with single and multiple viruses. Rotavirus vaccine was introduced in 2010, reaching a 70% coverage in 2 years. All rectal swabs collected from diarrheic children (<5 years) between December 2017 and March 2022 were included. Detection of the same viruses within 2 months was considered a single episode. Episodes with positive stool bacterial PCR were excluded. A total of 5879 samples were collected, revealing 86.9% (1509) with single virus detection and 13.1% (227) with multiple viruses. The most frequent combination was rotavirus and norovirus (27.8%), these infections followed a winter-spring seasonality akin to rotavirus. Children with multivirus infections exhibited higher immunodeficiency (OR 2.06) rates, but lower food allergy (OR 0.45) and prematurity rates (OR 0.55) compared to single infections. Greater disease severity, evaluated by the Vesikari score, was observed in multivirus episodes (p < 0.001, OR 1.12). Multivirus infections accounted for 13.1% of symptomatic cases in hospitalized young children. Despite vaccination efforts, rotavirus remained prominent, frequently in co-infections with norovirus. Overall, multivirus infections were linked to more severe diseases than single virus cases.
Collapse
Affiliation(s)
- Guy Hazan
- Pediatric Department D., Soroka University Medical Center, Faculty of Health Sciences, Ben Gurion University, Beer Sheva, Israel
- Pediatric Pulmonary Unit, Soroka University Medical Center, Faculty of Health Sciences, Ben Gurion University, Beer Sheva, Israel
| | - Yoav Goldstein
- Pediatric Department D., Soroka University Medical Center, Faculty of Health Sciences, Ben Gurion University, Beer Sheva, Israel
| | - David Greenberg
- Pediatric Infectious Diseases Unit, Soroka University Medical Center, Faculty of Health Sciences, Ben Gurion University, Beer Sheva, Israel
| | - Firas Khalde
- Pediatric Department D., Soroka University Medical Center, Faculty of Health Sciences, Ben Gurion University, Beer Sheva, Israel
| | - Rofaida Mahajna
- Pediatric Department D., Soroka University Medical Center, Faculty of Health Sciences, Ben Gurion University, Beer Sheva, Israel
| | - Ayelet Keren-Naos
- Clinical Virology Laboratory, Soroka University Medical Center, Beer Sheva, Israel
| | - Eli Hershkovitz
- Pediatric Department D., Soroka University Medical Center, Faculty of Health Sciences, Ben Gurion University, Beer Sheva, Israel
| | - Yaniv Faingelernt
- Pediatric Department D., Soroka University Medical Center, Faculty of Health Sciences, Ben Gurion University, Beer Sheva, Israel
- Pediatric Gastroenterology Hepatology and Nutrition Unit, Soroka University Medical Center, Faculty of Health Sciences, Ben Gurion University, Beer Sheva, Israel
| | - Noga Givon-Lavi
- Pediatric Infectious Diseases Unit, Soroka University Medical Center, Faculty of Health Sciences, Ben Gurion University, Beer Sheva, Israel
| | - Dana Danino
- Pediatric Department D., Soroka University Medical Center, Faculty of Health Sciences, Ben Gurion University, Beer Sheva, Israel
- Pediatric Infectious Diseases Unit, Soroka University Medical Center, Faculty of Health Sciences, Ben Gurion University, Beer Sheva, Israel
| |
Collapse
|
42
|
Poto R, Fusco W, Rinninella E, Cintoni M, Kaitsas F, Raoul P, Caruso C, Mele MC, Varricchi G, Gasbarrini A, Cammarota G, Ianiro G. The Role of Gut Microbiota and Leaky Gut in the Pathogenesis of Food Allergy. Nutrients 2023; 16:92. [PMID: 38201921 PMCID: PMC10780391 DOI: 10.3390/nu16010092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/21/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024] Open
Abstract
Food allergy (FA) is a growing public health concern, with an increasing prevalence in Western countries. Increasing evidence suggests that the balance of human gut microbiota and the integrity of our intestinal barrier may play roles in the development of FA. Environmental factors, including industrialization and consumption of highly processed food, can contribute to altering the gut microbiota and the intestinal barrier, increasing the susceptibility to allergic sensitization. Compositional and functional alterations to the gut microbiome have also been associated with FA. In addition, increased permeability of the gut barrier allows the translocation of allergenic molecules, triggering Th2 immune responses. Preclinical and clinical studies have highlighted the potential of probiotics, prebiotics, and postbiotics in the prevention and treatment of FA through enhancing gut barrier function and promoting the restoration of healthy gut microbiota. Finally, fecal microbiota transplantation (FMT) is now being explored as a promising therapeutic strategy to prevent FA in both experimental and clinical studies. In this review article, we aim to explore the complex interplay between intestinal permeability and gut microbiota in the development of FA, as well as depict potential therapeutic strategies.
Collapse
Affiliation(s)
- Remo Poto
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (R.P.); (G.V.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80131 Naples, Italy
| | - William Fusco
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy (E.R.); (M.C.); (F.K.); (P.R.); (C.C.); (M.C.M.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Medical and Surgical Sciences, UOSD DH Internal Medicine and Digestive Diseases, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Emanuele Rinninella
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy (E.R.); (M.C.); (F.K.); (P.R.); (C.C.); (M.C.M.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, Clinical Nutrition Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Marco Cintoni
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy (E.R.); (M.C.); (F.K.); (P.R.); (C.C.); (M.C.M.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, Clinical Nutrition Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Francesco Kaitsas
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy (E.R.); (M.C.); (F.K.); (P.R.); (C.C.); (M.C.M.); (A.G.); (G.C.)
| | - Pauline Raoul
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy (E.R.); (M.C.); (F.K.); (P.R.); (C.C.); (M.C.M.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, Clinical Nutrition Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Cristiano Caruso
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy (E.R.); (M.C.); (F.K.); (P.R.); (C.C.); (M.C.M.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, UOSD DH Internal Medicine and Digestive Diseases, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Maria Cristina Mele
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy (E.R.); (M.C.); (F.K.); (P.R.); (C.C.); (M.C.M.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, Clinical Nutrition Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (R.P.); (G.V.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80131 Naples, Italy
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy (E.R.); (M.C.); (F.K.); (P.R.); (C.C.); (M.C.M.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Medical and Surgical Sciences, UOSD DH Internal Medicine and Digestive Diseases, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Giovanni Cammarota
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy (E.R.); (M.C.); (F.K.); (P.R.); (C.C.); (M.C.M.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Medical and Surgical Sciences, UOSD DH Internal Medicine and Digestive Diseases, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Gianluca Ianiro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy (E.R.); (M.C.); (F.K.); (P.R.); (C.C.); (M.C.M.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Medical and Surgical Sciences, UOSD DH Internal Medicine and Digestive Diseases, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|
43
|
Arditi Z, Bunyavanich S. Commensal collaborations: Food allergy and the microbiome. J Allergy Clin Immunol 2023; 152:1417-1419. [PMID: 37558058 PMCID: PMC11402024 DOI: 10.1016/j.jaci.2023.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 07/18/2023] [Accepted: 08/03/2023] [Indexed: 08/11/2023]
Affiliation(s)
- Zoe Arditi
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Supinda Bunyavanich
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
44
|
Glatthardt T, van Tilburg Bernardes E, Arrieta MC. The mycobiome in atopic diseases: Inducers and triggers. J Allergy Clin Immunol 2023; 152:1368-1375. [PMID: 37865199 DOI: 10.1016/j.jaci.2023.10.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/03/2023] [Accepted: 10/17/2023] [Indexed: 10/23/2023]
Abstract
Atopic diseases are characterized by type 2 inflammation, with high levels of allergen-specific TH2 cell immune responses and elevated production of IgE. These common disorders have increased in incidence around the world, which is partly explained by detrimental disturbances to the early-life intestinal microbiome. Although most studies have focused exclusively on bacterial members of the microbiome, intestinal fungi have started to be recognized for their impact on host immune development and atopy pathogenesis. From this perspective, we review recent findings demonstrating the strong interactions between members of the mycobiome and the host immune system early in life, leading to immune tolerance during eubiosis or inducing sensitization and overt TH2 cell responses during dysbiosis. Current evidence places intestinal fungi as central players in the development of allergic diseases and potential targets for atopy prevention and treatments.
Collapse
Affiliation(s)
- Thais Glatthardt
- the Department of Physiology and Pharmacology, University of Calgary, Calgary; the International Microbiome Centre, Snyder Institute for Chronic Diseases, University of Calgary, Calgary; the Department of Pediatrics, Alberta Children Hospital Research Institute, University of Calgary, Calgary
| | - Erik van Tilburg Bernardes
- the Department of Physiology and Pharmacology, University of Calgary, Calgary; the International Microbiome Centre, Snyder Institute for Chronic Diseases, University of Calgary, Calgary; the Department of Pediatrics, Alberta Children Hospital Research Institute, University of Calgary, Calgary
| | - Marie-Claire Arrieta
- the Department of Physiology and Pharmacology, University of Calgary, Calgary; the International Microbiome Centre, Snyder Institute for Chronic Diseases, University of Calgary, Calgary; the Department of Pediatrics, Alberta Children Hospital Research Institute, University of Calgary, Calgary.
| |
Collapse
|
45
|
Chun Y, Grishin A, Rose R, Zhao W, Arditi Z, Zhang L, Wood RA, Burks AW, Jones SM, Leung DYM, Jones DR, Sampson HA, Sicherer SH, Bunyavanich S. Longitudinal dynamics of the gut microbiome and metabolome in peanut allergy development. J Allergy Clin Immunol 2023; 152:1569-1580. [PMID: 37619819 PMCID: PMC11440358 DOI: 10.1016/j.jaci.2023.08.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/20/2023] [Accepted: 08/09/2023] [Indexed: 08/26/2023]
Abstract
BACKGROUND Rising rates of peanut allergy (PA) motivate investigations of its development to inform prevention and therapy. Microbiota and the metabolites they produce shape food allergy risk. OBJECTIVE We sought to gain insight into gut microbiome and metabolome dynamics in the development of PA. METHODS We performed a longitudinal, integrative study of the gut microbiome and metabolome of infants with allergy risk factors but no PA from a multicenter cohort followed through mid-childhood. We performed 16S rRNA sequencing, short chain fatty acid measurements, and global metabolome profiling of fecal samples at infancy and at mid-childhood. RESULTS In this longitudinal, multicenter sample (n = 122), 28.7% of infants developed PA by mid-childhood (mean age 9 years). Lower infant gut microbiome diversity was associated with PA development (P = .014). Temporal changes in the relative abundance of specific microbiota and gut metabolite levels significantly differed in children who developed PA. PA-bound children had different abundance trajectories of Clostridium sensu stricto 1 sp (false discovery rate (FDR) = 0.015) and Bifidobacterium sp (FDR = 0.033), with butyrate (FDR = 0.045) and isovalerate (FDR = 0.036) decreasing over time. Metabolites associated with PA development clustered within the histidine metabolism pathway. Positive correlations between microbiota, butyrate, and isovalerate and negative correlations with histamine marked the PA-free network. CONCLUSION The temporal dynamics of the gut microbiome and metabolome in early childhood are distinct for children who develop PA. These findings inform our thinking on the mechanisms underlying and strategies for potentially preventing PA.
Collapse
Affiliation(s)
- Yoojin Chun
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Alexander Grishin
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Rebecca Rose
- Metabolomics Core, New York University, New York, NY
| | - William Zhao
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Zoe Arditi
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Lingdi Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Robert A Wood
- Department of Pediatrics, Johns Hopkins University, Baltimore, Md
| | - A Wesley Burks
- Department of Pediatrics, University of North Carolina, Chapel Hill, NC
| | - Stacie M Jones
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Ark
| | | | - Drew R Jones
- Metabolomics Core, New York University, New York, NY
| | - Hugh A Sampson
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Scott H Sicherer
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Supinda Bunyavanich
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
46
|
Abril AG, Carrera M, Pazos M. Immunomodulatory effect of marine lipids on food allergy. Front Nutr 2023; 10:1254681. [PMID: 38035353 PMCID: PMC10683508 DOI: 10.3389/fnut.2023.1254681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/24/2023] [Indexed: 12/02/2023] Open
Abstract
Seafood is highly enriched in n-3 long-chain polyunsaturated fatty acids (n-3 LCPUFAs), particularly eicosapentaenoic acid (EPA, 20:5 n-3) and docosahexaenoic acid (DHA, 22:6 n-3), in contrast to the ultra-processed foods included in the modern Western diet that have high levels of n-6 linoleic acid (LA, 18:2 n-6), precursor for the pro-inflammatory n-6 arachidonic acid (ARA, 20:4 n-6). The capacity of marine lipids to reduce plasmatic triglycerides and blood pressure have been well-described. Moreover, recent studies have also raised evidence of a potential regulatory action of marine lipids on inflammation, the immune system, and food allergy (FA). FA is considered one of the main concerns to become life threatening in food safety. The prevalence of this emerging global problem has been increasing during the last two decades, especially in industrialized countries. About a 6-8% of young children and 2-4% of adults is estimated to be affected by FA. The main objective of the current study is to update the existing knowledge, but also the limitations, on the potential impact of marine lipids and their lipid mediators in regulating immunity, inflammation, and ultimately, food allergies. In particular, the focus is on the effect of marine lipids in modulating the key factors that control the sensitization and effector phases of FA, including gut microbiota (GM), inflammation, and immune system response. Results in animal models highlight the positive effect that consuming marine lipids, whether as a supplement or through seafood consumption, may have a relevant role in improving gut dysbiosis and inflammation, and preventing or reducing the severity of FA. However, more systematic studies in humans are needed to optimize such beneficial actions to each particular FA, age, and medical condition to reach an effective clinical application of marine lipids to improve FAs and their outcomes.
Collapse
Affiliation(s)
- Ana G. Abril
- Department of Microbiology and Parasitology, Faculty of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, Spain
- Department of Food Technology, Institute of Marine Research (IM-CSIC), Spanish National Research Council (CSIC), Vigo, Spain
| | - Mónica Carrera
- Department of Food Technology, Institute of Marine Research (IM-CSIC), Spanish National Research Council (CSIC), Vigo, Spain
| | - Manuel Pazos
- Department of Food Technology, Institute of Marine Research (IM-CSIC), Spanish National Research Council (CSIC), Vigo, Spain
| |
Collapse
|
47
|
Zhang Q, Zhang C, Zhang Y, Liu Y, Wang J, Gao Z, Sun J, Li Q, Sun J, Cui X, Wang Y, Fu L. Early-life risk factors for food allergy: Dietary and environmental factors revisited. Compr Rev Food Sci Food Saf 2023; 22:4355-4377. [PMID: 37679957 DOI: 10.1111/1541-4337.13226] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 07/21/2023] [Accepted: 07/27/2023] [Indexed: 09/09/2023]
Abstract
There appears a steep increase in the prevalence of food allergy worldwide in the past few decades. It is believed that, rather than genetic factors, the recently altered dietary and environmental factors are the driving forces behind the rapid increase of this disease. Accumulating evidence has implied that external exposures that occurred in prenatal and postnatal periods could affect the development of oral tolerance in later life. Understanding the potential risk factors for food allergy would greatly benefit the progress of intervention and therapy. In this review, we present updated knowledge on the dietary and environmental risk factors in early life that have been shown to impact the development of food allergy. These predominantly include dietary habits, microbial exposures, allergen exposure routes, environmental pollutants, and so on. The key evidence, conflicts, and potential research topics of each theory are discussed, and associated interventional strategies to prevent the disease development and ameliorate treatment burden are included. Accumulating evidence has supported the causative role of certain dietary and environmental factors in the establishment of oral tolerance in early life, especially the time of introducing allergenic foods, skin barrier function, and microbial exposures. In addition to certain immunomodulatory factors, increasing interest is raised toward modern dietary patterns, where adequately powered studies are required to identify contributions of those modifiable risk factors. This review broadens our understanding of the connections between diet, environment, and early-life immunity, thus benefiting the progress of intervention and therapy of food allergy.
Collapse
Affiliation(s)
- Qiaozhi Zhang
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Chi Zhang
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Yong Zhang
- Nutrition Department of the First Medical Centre of PLA General Hospital, Beijing, China
| | - Yinghua Liu
- Nutrition Department of the First Medical Centre of PLA General Hospital, Beijing, China
| | - Jin Wang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, China
- Department of Bioresource Engineering, Faculty of Agricultural and Environmental Sciences, McGill University, Sainte-Anne-de-Bellevue, QC, Canada
| | - Zhongshan Gao
- Allergy Research Center, Zhejiang University, Hangzhou, China
| | - Jinlyu Sun
- Allergy Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qianqian Li
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Jiachen Sun
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Xin Cui
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Yanbo Wang
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Linglin Fu
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| |
Collapse
|
48
|
Yuan J, Tong P, Meng X, Wu Y, Li X, Gao J, Chen H. Oral exposure to Staphylococcus aureus enterotoxin B could promote the Ovalbumin-induced food allergy by enhancing the activation of DCs and T cells. Front Immunol 2023; 14:1250458. [PMID: 37908363 PMCID: PMC10615071 DOI: 10.3389/fimmu.2023.1250458] [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: 07/25/2023] [Accepted: 09/29/2023] [Indexed: 11/02/2023] Open
Abstract
Introduction Recent work highlighted the importance of environmental contaminants in the development of allergic diseases. Methods The intestinal mucosal barrier, Th (helper T) cells, DCs (dendritic cells), and intestinal flora were analyzed with flow cytometry, RNA-seq, and 16s sequencing in the present study to demonstrate whether the exposure of enterotoxins like Staphylococcus aureus enterotoxin B (SEB) in allergens could promote the development of food allergy. Results and discussion We found that co-exposure to SEB and Ovalbumin (OVA) could impair the intestinal barrier, imbalance the intestinal Th immune, and cause the decline of intestinal flora diversity in OVA-sensitized mice. Moreover, with the co-stimulation of SEB, the transport of OVA was enhanced in the Caco-2 cell monolayer, the uptake and presentation of OVA were promoted in the bone marrow dendritic cells (BMDCs), and Th cell differentiation was also enhanced. In summary, co-exposure to SEB in allergens should be considered a food allergy risk factor.
Collapse
Affiliation(s)
- Jin Yuan
- State Key Laboratory of Food Science and Resource, Nanchang University, Nanchang, China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, China
- College of Food Science & Technology, Nanchang University, Nanchang, China
| | - Ping Tong
- State Key Laboratory of Food Science and Resource, Nanchang University, Nanchang, China
| | - Xuanyi Meng
- Sino-German Joint Research Institute, Nanchang University, Nanchang, China
| | - Yong Wu
- Sino-German Joint Research Institute, Nanchang University, Nanchang, China
| | - Xin Li
- State Key Laboratory of Food Science and Resource, Nanchang University, Nanchang, China
- College of Food Science & Technology, Nanchang University, Nanchang, China
| | - Jinyan Gao
- College of Food Science & Technology, Nanchang University, Nanchang, China
| | - Hongbing Chen
- State Key Laboratory of Food Science and Resource, Nanchang University, Nanchang, China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, China
| |
Collapse
|
49
|
Scheurer S, Junker AC, He C, Schülke S, Toda M. The Role of IgA in the Manifestation and Prevention of Allergic Immune Responses. Curr Allergy Asthma Rep 2023; 23:589-600. [PMID: 37610671 PMCID: PMC10506939 DOI: 10.1007/s11882-023-01105-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2023] [Indexed: 08/24/2023]
Abstract
PURPOSE OF REVIEW Immunoglobulin A (IgA) mediates immune exclusion of antigens in the gut. Notably, IgA plays also a role in the prevention of IgE-mediated allergies and induction of immune tolerance. The present review addresses the role of IgA in the manifestation of IgE-mediated allergies, including allergen-specific immunotherapy (AIT), the regulation of IgA production, and the mechanism of IgA in immune cell activation. RECENT FINDINGS The majority of studies report an association of IgA with the induction of immune tolerance in IgE-mediated allergies. However, reports on the involvement of humoral and mucosal IgA, IgA subtypes, monomeric and polymeric IgA, and the mechanism of IgA-mediated immune cell activation are confounding. Effects by IgA are likely mediated by alteration of microbiota, IgE-blocking capacity, or activation of inhibitory signaling pathways. However, the precise mechanism of IgA-regulation, the contribution of serum and/or mucosal IgA, and IgA1/2 subtypes, on the manifestation of IgE-mediated allergies, and the underlying immune modulatory mechanism are still elusive.
Collapse
Affiliation(s)
- Stephan Scheurer
- Federal Institute for Vaccines and Biomedicines, Molecular Allergology, Paul-Ehrlich-Institut, Paul-Ehrlich Str., 51-58, 63225, Langen, Germany.
| | - Ann-Christine Junker
- Federal Institute for Vaccines and Biomedicines, Molecular Allergology, Paul-Ehrlich-Institut, Paul-Ehrlich Str., 51-58, 63225, Langen, Germany
| | - Chaoqi He
- Laboratory of Food and Biomolecular Science, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Stefan Schülke
- Federal Institute for Vaccines and Biomedicines, Molecular Allergology, Paul-Ehrlich-Institut, Paul-Ehrlich Str., 51-58, 63225, Langen, Germany
- Division of Allergology, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | - Masako Toda
- Laboratory of Food and Biomolecular Science, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| |
Collapse
|
50
|
Zhong H, Li J, Cheng JH. Targeting different signaling pathways for food allergy regulation and potential therapy: a review. Crit Rev Food Sci Nutr 2023; 64:12860-12877. [PMID: 37707435 DOI: 10.1080/10408398.2023.2257798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
The rising incidence rate of food allergy is attracting more intention. The pathogenesis of food allergy is complex and its definite regulatory mechanism is not utterly understood. Exploring the molecular mechanism of food allergy to help find effective methods that can prevent or treat food allergy is widely necessary. Recently, targeting cellular signaling pathways have been employed as novel approaches to discover food allergy therapy. Supplementing probiotics and bioactive compounds with anti-allergic property are believed feasible approaches for food allergy therapy. These probiotics or bioactive compounds affect food allergy by regulating cellular signaling pathways, and ultimately alleviate food allergy. This review aims to report systematic information about the knowledge of signaling pathways participated in food allergy, the alterations of these signaling pathways during food allergy that treated with probiotics and bioactive compounds are discussed as well. Further studies on the mechanism of signaling pathway network regulating food allergy and the precise action mechanism of probiotics and bioactive compounds are in the urgent need to help develop efficient treatment or complete prevention. We hope to help scientists understand food allergy systematically.
Collapse
Affiliation(s)
- Hangyu Zhong
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
- Academy of Contemporary Food Engineering, South China University of Technology, Guangzhou Higher Education Mega Centre, Guangzhou, China
| | - Jilin Li
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
- Academy of Contemporary Food Engineering, South China University of Technology, Guangzhou Higher Education Mega Centre, Guangzhou, China
| | - Jun-Hu Cheng
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
- Academy of Contemporary Food Engineering, South China University of Technology, Guangzhou Higher Education Mega Centre, Guangzhou, China
| |
Collapse
|