1
|
Li T, Zheng Q, Xu J, Li Y, Zhang M, Zhang B, Zhou L, Tian J. Comparison of 11 Formulas and Breastfeeding for Atopic Dermatitis and Growth in Pediatric Cow's Milk Protein Allergy: A Systematic Review and Network Meta-Analysis of 23 Randomized Controlled Trials. J Evid Based Med 2025; 18:e70026. [PMID: 40178916 DOI: 10.1111/jebm.70026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 02/20/2025] [Accepted: 03/20/2025] [Indexed: 04/05/2025]
Abstract
OBJECTIVE This study aimed to evaluate the effectiveness of various formulas and the ability of breastfeeding with the exclusion of cow milk protein to reduce the Scoring Atopic Dermatitis (SCORAD) index and promote growth in infants with cow milk protein allergy. METHODS We conducted a systematic search of PubMed, Embase, Web of Science, the Cochrane Central Register of Controlled Trials (CENTRAL), ClinicalTrials.gov, China National Knowledge Infrastructure, WanFang Data, Weipu, and the China Biomedical Literature Database. The search period ranged from the inception of each database to December 2023 (with an update until January 15, 2025). We included randomized controlled trials (RCTs) comparing formulas and breastfeeding for cow's milk protein allergy in infants. Two independent reviewers extracted data via standardized methods and assessed the risk of bias via the revised Cochrane risk-of-bias 2.0 tool. We performed a network meta-analysis (NMA) via a Bayesian fixed-effects model in RStudio and assessed the certainty of the evidence via the Confidence in Network Meta-Analysis (CINeMA) online application. The protocol for this NMA was preregistered in PROSPERO (No. CRD42024504707). RESULTS This analysis included 23 RCTs involving 1997 children and assessed 12 interventions. Compared with the regular formula, the pectin-thickened amino acid formula (TAAF) might reduce the SCORAD index (-12.49, 95% confidence interval [CI] -20.38 to -4.48, low certainty). At ≤6 months of follow-up, compared with rice-hydrolyzed formula (RHF), breastfeeding might improve the length-for-age Z score (LAZ) (0.47, 95% CI 0.13-0.81, moderate certainty), and breastfeeding (0.39, 95% CI 0.02-0.77, low certainty) and extensively hydrolyzed formula (EHF) with probiotics (0.38, 95% CI 0.00-0.77, low certainty) might respectively improve the weight-for-age Z score (WAZ) and weight-for-length Z score (WLZ). At the 12-month follow-up, EHF might improve the LAZ (0.41, 95% CI 0.11-0.71, low certainty) and WLZ (0.37, 95% CI 0.18-0.56, low certainty) compared with RHF, whereas the amino acid formula (AAF) may improve the WAZ (0.33, 95% CI 0.02-0.63, low certainty). CONCLUSIONS Low-certainty evidence suggested that TAAF might reduce the SCORAD index. Moderate or low certainty evidence indicated that, at ≤6 months of follow-up, breastfeeding might improve the LAZ and WAZ, whereas EHF with probiotics might improve the WLZ. At the 12-month follow-up, EHF might improve the LAZ and WLZ, whereas AAF might improve the WAZ. However, further high-quality studies would be needed to confirm these findings and assess their safety and cost-effectiveness.
Collapse
Affiliation(s)
- Tengfei Li
- School of Nursing, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Evidence Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Qingyong Zheng
- Evidence Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Jianguo Xu
- Evidence Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Yiyi Li
- School of Nursing, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Mingyue Zhang
- Evidence Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Bowa Zhang
- School of Nursing, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Li Zhou
- Department of Pediatric Gastroenterology, Gansu Province Maternity and Child Health Hospital, Gansu Province Central Hospital, Lanzhou, Gansu, China
| | - Jinhui Tian
- Evidence Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
2
|
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
|
3
|
Jiang L, Zhang L, Xia J, Cheng L, Chen G, Wang J, Raghavan V. Probiotics supplementation during pregnancy or infancy on multiple food allergies and gut microbiota: a systematic review and meta-analysis. Nutr Rev 2025; 83:e25-e41. [PMID: 38502006 PMCID: PMC11723154 DOI: 10.1093/nutrit/nuae024] [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] [Indexed: 03/20/2024] Open
Abstract
CONTEXT Probiotics show promise in preventing and managing food allergies, but the impact of supplementation during pregnancy or infancy on children's allergies and gut microbiota remains unclear. OBJECTIVE This study aimed to assess the effects of maternal or infant probiotic supplementation on food allergy risk and explore the role of gut microbiota. DATA SOURCES A systematic search of databases (PubMed, Cochrane Library, Embase, and Medline) identified 37 relevant studies until May 20, 2023. DATA EXTRACTION Two independent reviewers extracted data, including probiotics intervention details, gut microbiota analysis, and food allergy information. DATA ANALYSIS Probiotics supplementation during pregnancy and infancy reduced the risk of total food allergy (relative risk [RR], 0.79; 95% CI, 0.63-0.99), cow-milk allergy (RR, 0.51; 95% CI, 0.29-0.88), and egg allergy (RR, 0.57; 95% CI, 0.39-0.84). Infancy-only supplementation lowered cow-milk allergy risk (RR, 0.69; 95% CI, 0.49-0.96), while pregnancy-only had no discernible effect. Benefits were observed with over 2 probiotic species, and a daily increase of 1.8 × 109 colony-forming units during pregnancy and infancy correlated with a 4% reduction in food allergy risk. Children with food allergies had distinct gut microbiota profiles, evolving with age. CONCLUSIONS Probiotics supplementation during pregnancy and infancy reduces food allergy risk and correlates with age-related changes in gut microbial composition in children. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CRD42023425988.
Collapse
Affiliation(s)
- Lan Jiang
- 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
| | - Lili Zhang
- 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
| | - Jiayue Xia
- 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
| | - Lei Cheng
- Department of Otorhinolaryngology and Clinical Allergy Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Guoxun Chen
- Department of Nutrition, University of Tennessee at Knoxville, Knoxville, TN, USA
| | - 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
| | - Vijaya Raghavan
- Department of Bioresource Engineering, Faculty of Agricultural and Environmental Sciences, McGill University, Sainte-Anne-de-Bellevue, QC, Canada
| |
Collapse
|
4
|
Tan-Lim CSC, Esteban-Ipac NAR. Systematic review and meta-analysis on probiotics as treatment for food allergies among pediatric patients: A 2024 update. Pediatr Allergy Immunol 2025; 36:e70028. [PMID: 39803979 DOI: 10.1111/pai.70028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 12/15/2024] [Accepted: 01/03/2025] [Indexed: 05/02/2025]
Abstract
This systematic review updated the available evidence on the effectiveness and safety of probiotics as treatment of food allergy among pediatric patients. We conducted a systematic search for all randomized controlled trials available until March 13, 2024 that evaluated the effectiveness and safety of probiotics for treating pediatric food allergy. Two authors independently conducted the search, screening, and data extraction. Data analysis and synthesis were done using Review Manager 5.4 software. We included 13 articles involving 1608 pediatric patients with food allergy. Probiotics probably has no effect on reducing eczema scores among infants with CMA (MD -1.29 points, 95% CI -4.14, 1.56; moderate certainty of evidence), based on two studies. Probiotics may reduce eczema scores for children with various types of allergy (MD -23.08 points, 95% CI -27.55, -18.61; low certainty of evidence), based on one study. It is uncertain whether probiotics may lead to tolerance acquisition among infants with CMA (RR 0.58, 95% CI 0.34, 1.00) due to very low certainty of evidence. Subgroup analysis based on time period showed significant benefit in inducing tolerance after at least 2 years (RR 0.44, 95% CI 0.29, 0.67; moderate certainty of evidence), suggesting a duration-dependent effect of probiotic usage. Subgroup analysis by probiotic preparation showed significant benefit for the LGG strain (RR 0.41, 95% CI 0.28, 0.62). Probiotics were generally well tolerated by the study participants. Further well-designed RCTs focusing on specific types of food allergy, as well as the use of standardized probiotic strains, outcome measurement, and longer follow-up periods are needed to draw clinically relevant conclusions on the role of probiotics in treating children with food allergy.
Collapse
Affiliation(s)
- Carol Stephanie C Tan-Lim
- Department of Clinical Epidemiology, College of Medicine, University of the Philippines Manila, Manila, Philippines
| | | |
Collapse
|
5
|
Lamminpää I, Niccolai E, Amedei A. Probiotics as adjuvants to mitigate adverse reactions and enhance effectiveness in Food Allergy Immunotherapy. Scand J Immunol 2024; 100:e13405. [PMID: 39407442 DOI: 10.1111/sji.13405] [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: 12/04/2023] [Revised: 08/04/2024] [Accepted: 08/18/2024] [Indexed: 11/21/2024]
Abstract
In the past decades, food allergies became increasingly dominant since early childhood, leading to a lower quality of life and to increasing costs addressed by the health care system. Beside standard avoidance of specific allergens and drug treatments following allergen exposure, a great deal of research has lately focused on Food Allergy Allergen Immunotherapy (FA-AIT). SCIT and EPIT (Subcutaneous and Epicutaneous Immunotherapy), OIT (Oral Immunotherapy), and SLIT (Sublingual Immunotherapy) consist in gradual exposure to allergens to desensitize and achieve tolerance once therapy has ended. Although promising, FA-AIT may bring acute local and systemic adverse reactions. To enhance efficacy, safety and convenience of AIT, the quest of potential adjuvants to mitigate the adverse reactions becomes crucial. Immunomodulatory activities, such as that of increasing the regulatory T cells and decreasing the IgE, have been observed in specific probiotics' strains and multiple studies elucidated the role of gut microbiota as a major interplayer among the host and its immune system. In this review, the microbiome modulation is shown as potential AIT adjuvant, nevertheless the need of more clinical studies in the near future is pivotal to assess the efficacy of targeted bacterial therapies and faecal microbiota transplantation.
Collapse
Affiliation(s)
- Ingrid Lamminpää
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
- SOD of Interdisciplinary Internal Medicine, Azienda Ospedaliera Universitaria Careggi (AOUC), Florence, Italy
| | - Elena Niccolai
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
- SOD of Interdisciplinary Internal Medicine, Azienda Ospedaliera Universitaria Careggi (AOUC), Florence, Italy
| | - Amedeo Amedei
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
- SOD of Interdisciplinary Internal Medicine, Azienda Ospedaliera Universitaria Careggi (AOUC), Florence, Italy
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Florence, Italy
| |
Collapse
|
6
|
Bognanni A, Firmino RT, Arasi S, Chu DK, Chu AW, Waffenschmidt S, Agarwal A, Dziechciarz P, Horvath A, Mihara H, Roldan Y, Terracciano L, Martelli A, Starok A, Said M, Shamir R, Ansotegui IJ, Dahdah L, Ebisawa M, Galli E, Kamenwa R, Lack G, Li H, Pawankar R, Warner A, Wong GWK, Bozzola M, Assa'Ad A, Dupont C, Bahna S, Spergel J, Venter C, Szajewska H, Nowak-Wegrzyn AH, Vandenplas Y, Papadopoulos NG, Waserman S, Fiocchi A, Schünemann HJ, Brożek JL. World Allergy Organization (WAO) Diagnosis and Rationale for Action against Cow's Milk Allergy (DRACMA) guideline update - XI - Milk supplement/replacement formulas for infants and toddlers with CMA - Systematic review. World Allergy Organ J 2024; 17:100947. [PMID: 39310372 PMCID: PMC11415968 DOI: 10.1016/j.waojou.2024.100947] [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: 02/15/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 09/25/2024] Open
Abstract
Background Cow's milk allergy (CMA) is the most complex and common food allergy in infants. Elimination of cow's milk from the diet and replacement with a specialized formula for infants with cow's milk allergy who cannot be breastfed is an established approach to minimize the risk of severe allergic reactions while avoiding nutritional deficiencies. Given the availability of multiple options, such as extensively hydrolyzed cow's milk-based formula (eHF-CM), aminoacid formula (AAF), hydrolyzed rice formula (HRF), and soy formula (SF), there is some uncertainty regarding which formula might represent the most suitable choice with respect to health outcomes. The addition of probiotics to a specialized formula has also been proposed as a potential approach to possibly increase the benefit. We systematically reviewed specialized formulas for infants with CMA to inform the updated World Allergy Organization (WAO) DRACMA guidelines. Objective To systematically review and synthesize the available evidence about the use of specialized formulas for the management of individuals with CMA. Methods We searched from inception PubMed, Medline, Embase, the Cochrane Central Register of Controlled Trials (CENTRAL), and the websites of selected allergy organizations, for randomized and non-randomized trials of any language investigating specialized formulas with or without probiotics. We included all studies irrespective of the language of the original publication. The last search was conducted in January 2024. We synthesized the identified evidence quantitatively or narratively as appropriate and summarized it in the evidence profiles. We conducted this review following the PRISMA, Cochrane methods, and the GRADE approach. Results We identified 3558 records including 14 randomized trials and 7 observational studies. Very low certainty evidence suggested that in infants with IgE-mediated CMA, eHF-CM, compared with AAF, might have higher probability of outgrowing CMA (risk ratio (RR) 2.32; risk difference (RD) 25 more per 100), while showing potentially lower probability of severe vomiting (RR 0.12, 95% CI 0.02 to 0.88; RD 23 fewer per 100, 95% CI 3 to 26) and developing food protein-induced enterocolitis syndrome (FPIES) (RR 0.15, 95% CI 0.03 to 0.82; RD 34 fewer per 100, 95% CI 7 to 39). We also found, however, that eHF-CM might be inferior to AAF in supporting a physiological growth, with respect to both weight (-5.5% from baseline, 95%CI -9.5% to -1.5%) and length (-0.7 z-score change, 95%CI -1.15 to -0.25) (very low certainty). We found similar effects for eHF-CM, compared with AAF, also in non-IgE CMA. When compared with SF, eHF-CM might favor weight gain for IgE CMA infants (0.23 z-score change, 95%CI 0.01 to 0.45), and tolerance acquisition (RR 1.86, 95%CI 1.03 to 3.37; RD 27%, 95%CI 1%-74%) for non-IgE CMA (both at very low certainty of the evidence (CoE)). The comparison of eHF-CM vs. HRF, and HRF vs. SF, showed no difference in effect (very low certainty). For IgE CMA patients, low certainty evidence suggested that adding probiotics (L. rhamnosus GG, L. casei CRL431 and B. lactis Bb-12) might increase the probability of developing CMA tolerance (RR 2.47, 95%CI 1.03 to 5.93; RD 27%, 95%CI 1%-91%), and reduce the risk of severe wheezing (RR 0.12, 95%CI 0.02 to 0.95; RD -23%, 95%CI -8% to -0.4%). However, in non-IgE CMA infants, the addition of probiotics (L. rhamnosus GG) showed no significant effect, as supported by low to very low CoE. Conclusions Currently available studies comparing eHF-CM, AAF, HRF, and SF provide very low certainty evidence about their effects in infants with IgE-mediated and non-IgE-mediated CMA. Our review revealed several limitations in the current body of evidence, primarily arising from concerns related to the quality of studies, the limited size of the participant populations and most importantly the lack of diversity and standardization in the compared interventions. It is therefore imperative for future studies to be methodologically rigorous and investigate a broader spectrum of available interventions. We encourage clinicians and researchers to review current World Allergy Organization (WAO) Diagnosis and Rationale for Action against Cow's Milk Allergy (DRACMA) Guidelines for suggestions on how to use milk replacement formulas in clinical practice and what additional research would be the most beneficial.
Collapse
Affiliation(s)
- Antonio Bognanni
- Department of Health Research Methods, Evidence & Impact, McMaster University, Hamilton, Ontario, Canada
- Clinical Epidemiology and Research Center (CERC), Humanitas University & Humanitas Research Hospital, Pieve Emanuele, Milano, Italy
- Department of Medicine, Evidence in Allergy Group, McMaster University, Hamilton, Ontario, Canada
| | - Ramon T. Firmino
- Academic Unit of Biological Sciences, Federal University of Campina Grande, Patos, Paraíba, Brazil
| | - Stefania Arasi
- Translational Research in Pediatric Specialties Area, Division of Allergy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Derek K. Chu
- Department of Health Research Methods, Evidence & Impact, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, Evidence in Allergy Group, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, Division of Clinical Immunology and Allergy, McMaster University, Hamilton, Ontario, Canada
| | - Alexandro W.L. Chu
- Department of Medicine, Evidence in Allergy Group, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Siw Waffenschmidt
- Institute for Quality and Efficiency in Health Care, Cologne, Germany
| | - Arnav Agarwal
- Department of Medicine, Division of Internal Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | | | - Hanako Mihara
- Clinical Development Infectious Disease, Moderna Japan Co., Ltd., Japan
| | | | - Luigi Terracciano
- Pediatric Primary Care, National Pediatric Health Care System, Milan, Italy
- Italian Society of Preventive and Social Pediatrics (SIPPS), Italy
| | - Alberto Martelli
- Italian Society of Allergy and Pediatric Immunology (SIAIP), Italy
| | | | - Maria Said
- Allergy & Anaphylaxis Australia, Sydney, Australia
| | - Raanan Shamir
- Institute for Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center of Israel, Faculty of Medical and Health Sciences, Tel Aviv University, Israel
| | - Ignacio J. Ansotegui
- Department of Allergy & Immunology, Hospital Quironsalud Bizkaia, Erandio, Bilbao, Spain
| | - Lamia Dahdah
- Translational Research in Pediatric Specialties Area, Division of Allergy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Motohiro Ebisawa
- Clinical Research Center for Allergy and Rheumatology, NHO Sagamihara National Hospital, Kanagawa, Japan
| | - Elena Galli
- Pediatric Allergy Unit, Research Center, San Pietro Hospital - Fatebenefratelli, Rome, Italy
| | - Rose Kamenwa
- Department of Pediatrics and Child Health, Aga Khan University Hospital, Nairobi, Kenya
| | - Gideon Lack
- Department of Women and Children's Health/Peter Gorer Department of Immunobiology, School of Life Course Sciences, Faculty of Life Sciences & Medicine, King's College London, United Kingdom
- Evelina London Children's Hospital, Guy's and St Thomas' Hospital NHS Foundation Trust, United Kingdom
| | - Haiqi Li
- Department of Primary Child Care, Children's Hospital, Chongqing Medical University, China
| | - Ruby Pawankar
- Department of Pediatrics. Nippon Medical School, Tokyo, Japan
| | | | - Gary Wing Kin Wong
- Department of Paediatrics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Martin Bozzola
- Pediatric Allergy and Immunology Section, Dept of Pediatrics, Hospital Británico de Buenos Aires, Buenos Aires, Argentina
| | - Amal Assa'Ad
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | | | - Sami Bahna
- Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, USA
| | - Jonathan Spergel
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, USA
- Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA, USA
| | - Carina Venter
- Section of Allergy and Clinical Immunology, University of Colorado, USA
- Children's Hospital Colorado. Denver, Colorado, USA
| | - Hania Szajewska
- Department of Paediatrics, The Medical University of Warsaw, Warsaw, Poland
| | - Anna H. Nowak-Wegrzyn
- Department of Pediatrics, Hassenfeld Children's Hospital, New York University, Grossman School of Medicine, New York, NY, USA
- Department of Pediatrics, Gastroenterology and Nutrition, Collegium Medicum, University of Warmia and Mazury, Olsztyn, Poland
| | - Yvan Vandenplas
- Vrije Universiteit Brussel (VUB), UZ Brussel, Belgium
- KidZ Health Castle, Brussels, Belgium
| | - Nikolaos G. Papadopoulos
- Allergy Department, 2nd Pediatric Clinic, University of Athens, Athens, Greece
- Lydia Becker Institute, University of Manchester, Manchester, United Kingdom
| | - Susan Waserman
- Department of Medicine, Division of Clinical Immunology and Allergy, McMaster University, Hamilton, Ontario, Canada
| | - Alessandro Fiocchi
- Translational Research in Pediatric Specialties Area, Division of Allergy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Holger J. Schünemann
- Clinical Epidemiology and Research Center (CERC), Humanitas University & Humanitas Research Hospital, Pieve Emanuele, Milano, Italy
| | - Jan L. Brożek
- Department of Health Research Methods, Evidence & Impact, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, Division of Clinical Immunology and Allergy, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
7
|
Hesser LA, Puente AA, Arnold J, Ionescu E, Mirmira A, Talasani N, Lopez J, Maccio-Maretto L, Mimee M, Nagler CR. A synbiotic of Anaerostipes caccae and lactulose prevents and treats food allergy in mice. Cell Host Microbe 2024; 32:1163-1176.e6. [PMID: 38906158 PMCID: PMC11239278 DOI: 10.1016/j.chom.2024.05.019] [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: 07/20/2023] [Revised: 03/26/2024] [Accepted: 05/28/2024] [Indexed: 06/23/2024]
Abstract
Depletion of beneficial microbes by modern lifestyle factors correlates with the rising prevalence of food allergies. Re-introduction of allergy-protective bacteria may be an effective treatment strategy. We characterized the fecal microbiota of healthy and food-allergic infants and found that the anaerobe Anaerostipes caccae (A. caccae) was representative of the protective capacity of the healthy microbiota. We isolated a strain of A. caccae from the feces of a healthy infant and identified lactulose as a prebiotic to optimize butyrate production by A. caccae in vitro. Administration of a synbiotic composed of our isolated A. caccae strain and lactulose increased luminal butyrate in gnotobiotic mice colonized with feces from an allergic infant and in antibiotic-treated specific pathogen-free (SPF) mice, and prevented or treated an anaphylactic response to allergen challenge. The synbiotic's efficacy in two models and microbial contexts suggests that it may be a promising approach for the treatment of food allergy.
Collapse
Affiliation(s)
- Lauren A Hesser
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, USA
| | - Armando A Puente
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, USA
| | - Jack Arnold
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, USA
| | - Edward Ionescu
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, USA
| | - Anjali Mirmira
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Nidhi Talasani
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Jacqueline Lopez
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | | | - Mark Mimee
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, USA; Committee on Microbiology, The University of Chicago, Chicago, IL, USA
| | - Cathryn R Nagler
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, USA; Department of Pathology, The University of Chicago, Chicago, IL, USA; Committee on Immunology, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
8
|
Carucci L, Coppola S, Carandente R, Canani RB. Targeting Food Allergy with Probiotics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1449:79-93. [PMID: 39060732 DOI: 10.1007/978-3-031-58572-2_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
The globally dramatic increase in food allergy prevalence and severity is demanding effective preventive and therapeutic strategies. Food allergy derives from a defect of immune tolerance mechanisms. Immune tolerance is modulated by gut microbiome composition and function, and gut microbiome dysbiosis has been associated with the development of food allergy. Selected probiotic strains could regulate immune tolerance mechanisms. The mechanisms are multiple and are still not completely defined. Increasing evidence is providing useful information on the choice of optimal bacterial species/strains, dosage, and timing for intervention. The increased knowledge on the crucial role played by postbiotic gut microbiome-derived metabolites, such as butyrate, is also opening the way to a post- biotic approach in the stimulation of immune tolerance.
Collapse
Affiliation(s)
- Laura Carucci
- Department of Translational Medical Science at the University of Naples Federico II, Naples, Italy
| | - Serena Coppola
- Department of Translational Medical Science at the University of Naples Federico II, Naples, Italy
| | - Rosilenia Carandente
- Department of Translational Medical Science at the University of Naples Federico II, Naples, Italy
| | - Roberto Berni Canani
- Department of Translational Medical Science at the University of Naples Federico II, Naples, Italy.
| |
Collapse
|
9
|
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: 11] [Impact Index Per Article: 5.5] [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
|
10
|
Jasielska M, Buczyńska A, Adamczyk P, Grzybowska-Chlebowczyk U. Nutritional Status of Children with Newly Diagnosed Food Allergies. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1687. [PMID: 37892350 PMCID: PMC10605198 DOI: 10.3390/children10101687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/28/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023]
Abstract
BACKGROUND Most published pediatric guidelines on food allergy highlight the importance of nutritional counseling and dietary adequacy to avoid either growth retardation or nutritional deficiencies. The aim of the study was an assessment of the nutritional status of children with IgE-mediated food allergies. MATERIAL AND METHOD 45 patients with newly diagnosed food allergy (FA) and 33 healthy controls were analyzed (aged 6 to 72 months, 60.2% boys). The nutritional status was assessed using anthropometric measurements (body weight and length) and serum laboratory tests. The results were analyzed with the Statistica 12 software (Tulsa, OK, USA). RESULTS 82%, 40%, 8.8%, and 6.6% of the studied children demonstrated allergy to hen's egg, cow's milk, pork meat, and wheat/rye, respectively. Z-score BMI < -2SD was more often found in the FA subjects under 30 months of age than in the controls (p = 0.04). As many as 77.8% of the FA subjects and 78.8% of the controls were of normal height (hSDS: -0.23 ± 1.74 and -0.31 ± 1.49, respectively, p = 0.8). Retinol binding protein four serum concentration was significantly lower in the FA group (17.01 ± 3.84 mg/L) than in the controls (20.47 ± 4.87 mg/L, p < 0.001). No statistically significant differences were observed between the FA group and the controls (either in the younger or the older age group) (p > 0.05) for the serum concentrations of total protein, total cholesterol, thyroxin-binding prealbumin (TBPA), 25(OH)D, hemoglobin level or white blood cells. CONCLUSIONS In patients under 30 months of age, one of the symptoms of food allergy may be body weight deficiency, while short stature is less common at the time of diagnosis.
Collapse
Affiliation(s)
- Martyna Jasielska
- Department of Pediatrics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland; (A.B.); (P.A.)
| | | | | | | |
Collapse
|
11
|
Strisciuglio C, Vitale A, Perna F, Garziano F, Dolce P, Vitale S, Micillo T, Oglio F, Del Giudice MM, Matarese G, Gianfrani C. Bifidobacteria modulate immune response in pediatric patients with cow's milk protein allergy. Pediatr Res 2023; 94:1111-1118. [PMID: 36959319 DOI: 10.1038/s41390-023-02534-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 01/15/2023] [Indexed: 03/25/2023]
Abstract
BACKGROUND In children with an allergy to cow's milk proteins (CMA), the altered composition of intestinal microbiota influences the immune tolerance to milk proteins (CMP). This study aims to investigate the effect of probiotics on the phenotype and activation status of peripheral basophils and lymphocytes in a pediatric CMA cohort. METHODS CMA children underwent 45 days of treatment with Bifidobacteria. The basophil degranulation and the immune phenotype of B cells, T helper cells, and regulatory T cells were analyzed in peripheral blood at diagnosis (T0), after a 45-day probiotic treatment (T1), and 45 days after the probiotic wash-out (T2). RESULTS We observed in probiotic-treated CMA patients a decrease in naive T lymphocytes. Among the CD3+ cell subsets, both naive and activated CD4+ cells resulted markedly reduced after taking probiotics, with the lowest percentages at T2. A decreased basophil degranulation was observed in response to all analyzed CMP at T1 compared to T0. CONCLUSIONS The probiotic treatment resulted in a decrease of circulating naive and activated CD4+ T cells, as well as degranulating basophils. These data suggest that the Bifidobacteria could have a beneficial effect in the modulation of oral tolerance to CMP. TRIAL REGISTRATION ISRCTN69069358. URL of registration: https://www.isrctn.com/ISRCTN69069358 . IMPACT Probiotic treatment with Bifidobacteria induces a reduction of both naive and activated circulating CD4+ T cells in pediatric patients with cow's milk allergy (CMA). The probiotic supplementation induces a decreased basophil degranulation. The immunological tolerance persists even after 45 days of the probiotic wash-out. Bifidobacteria in vivo supplementation down-modulates the activation of innate and adaptive immunity in pediatric patients with cow's milk allergy. Bifidobacteria contribute to the development of immune tolerance in CMA patients.
Collapse
Affiliation(s)
- Caterina Strisciuglio
- Department of Woman, Child and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy.
| | - Alessandra Vitale
- Department of Woman, Child and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Francesco Perna
- Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy
| | - Federica Garziano
- U.O.C Clinical Biochemistry, A.O.R.N. Ospedale dei Colli, Naples, Italy
| | - Pasquale Dolce
- Department of Public Health, University of Naples "Federico II", Naples, Italy
| | - Serena Vitale
- Institute of Biochemistry and Cell Biology, CNR, Naples, Italy
| | - Teresa Micillo
- Neuroimmunology Unit, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Franca Oglio
- Department of Translational Medical Science, Section of Pediatrics, University of Naples "Federico II", Naples, Italy
| | - Michele Miraglia Del Giudice
- Department of Woman, Child and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giuseppe Matarese
- Department of Molecular Medicine and Biotechnology Science, University of Naples "Federico II", Naples, Italy
| | | |
Collapse
|
12
|
Cela L, Brindisi G, Gravina A, Pastore F, Semeraro A, Bringheli I, Marchetti L, Morelli R, Cinicola B, Capponi M, Gori A, Pignataro E, Piccioni MG, Zicari AM, Anania C. Molecular Mechanism and Clinical Effects of Probiotics in the Management of Cow's Milk Protein Allergy. Int J Mol Sci 2023; 24:9781. [PMID: 37372929 DOI: 10.3390/ijms24129781] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/23/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
Cow's milk protein allergy (CMPA) is the most common food allergy (FA) in infancy, affecting approximately 2% of children under 4 years of age. According to recent studies, the increasing prevalence of FAs can be associated with changes in composition and function of gut microbiota or "dysbiosis". Gut microbiota regulation, mediated by probiotics, may modulate the systemic inflammatory and immune responses, influencing the development of allergies, with possible clinical benefits. This narrative review collects the actual evidence of probiotics' efficacy in the management of pediatric CMPA, with a specific focus on the molecular mechanisms of action. Most studies included in this review have shown a beneficial effect of probiotics in CMPA patients, especially in terms of achieving tolerance and improving symptoms.
Collapse
Affiliation(s)
- Ludovica Cela
- Department of Maternal Infantile and Urological Science, Sapienza University of Rome, 00161 Rome, Italy
| | - Giulia Brindisi
- Department of Maternal Infantile and Urological Science, Sapienza University of Rome, 00161 Rome, Italy
| | - Alessandro Gravina
- Department of Maternal Infantile and Urological Science, Sapienza University of Rome, 00161 Rome, Italy
| | - Francesca Pastore
- Department of Maternal Infantile and Urological Science, Sapienza University of Rome, 00161 Rome, Italy
| | - Antonio Semeraro
- Department of Maternal Infantile and Urological Science, Sapienza University of Rome, 00161 Rome, Italy
| | - Ivana Bringheli
- Department of Maternal Infantile and Urological Science, Sapienza University of Rome, 00161 Rome, Italy
| | - Lavinia Marchetti
- Department of Maternal Infantile and Urological Science, Sapienza University of Rome, 00161 Rome, Italy
| | - Rebecca Morelli
- Department of Maternal Infantile and Urological Science, Sapienza University of Rome, 00161 Rome, Italy
| | - Bianca Cinicola
- Department of Maternal Infantile and Urological Science, Sapienza University of Rome, 00161 Rome, Italy
| | - Martina Capponi
- Department of Maternal Infantile and Urological Science, Sapienza University of Rome, 00161 Rome, Italy
| | - Alessandra Gori
- Department of Maternal Infantile and Urological Science, Sapienza University of Rome, 00161 Rome, Italy
| | - Elia Pignataro
- Department of Maternal Infantile and Urological Science, Sapienza University of Rome, 00161 Rome, Italy
| | - Maria Grazia Piccioni
- Department of Maternal Infantile and Urological Science, Sapienza University of Rome, 00161 Rome, Italy
| | - Anna Maria Zicari
- Department of Maternal Infantile and Urological Science, Sapienza University of Rome, 00161 Rome, Italy
| | - Caterina Anania
- Department of Maternal Infantile and Urological Science, Sapienza University of Rome, 00161 Rome, Italy
| |
Collapse
|
13
|
Ćesić D, Lugović Mihić L, Ozretić P, Lojkić I, Buljan M, Šitum M, Zovak M, Vidović D, Mijić A, Galić N, Tambić Andrašević A. Association of Gut Lachnospiraceae and Chronic Spontaneous Urticaria. Life (Basel) 2023; 13:1280. [PMID: 37374063 PMCID: PMC10301119 DOI: 10.3390/life13061280] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
(1) Background: Chronic spontaneous urticaria (CSU) has been linked to the dysbiosis of the gut microbiota. Furthermore, various studies have highlighted the anti-inflammatory properties of short-chain fatty acids (SCFAs), whose production is primarily regulated by the gut microbiota. However, only a few studies have investigated the role of major SCFA producers, such as Lachnospiraceae, in skin inflammatory diseases. (2) Goal: This study aimed to compare the abundance of Lachnospiraceae between CSU patients and healthy controls (HCs). (3) Material and methods: In this case-control study, 16S rRNA sequencing was performed to compare the composition of the gut microbiome between 22 CSU patients and 23 HCs. (4) Results: Beta-diversity revealed significant clustering (p < 0.05) between the CSU patients and HCs. Alpha diversity in the CSU group was significantly decreased according to the Evenness index (p < 0.05). The linear discriminant analysis effect size (LEfSe) identified the significant depletion of the Lachnospiraceae family in CSU patients. (5) Conclusion: Our study revealed the dysbiosis of the gut microbiota in CSU patients, including decreased levels of Lachnospiraceae members, responsible for SCFA production, suggesting that SCFAs may contribute to immune dysfunction in the pathogenesis of CSU. We speculate that the modulation of SCFAs could serve as a prospective additional option in CSU treatment.
Collapse
Affiliation(s)
- Diana Ćesić
- Department of Dermatology and Venereology, Medikol Clinic, 10000 Zagreb, Croatia
- School of Dental Medicine, University of Zagreb, 10000 Zagreb, Croatia; (L.L.M.); (M.B.); (M.Š.); (M.Z.); (D.V.); (A.M.); (N.G.); (A.T.A.)
| | - Liborija Lugović Mihić
- School of Dental Medicine, University of Zagreb, 10000 Zagreb, Croatia; (L.L.M.); (M.B.); (M.Š.); (M.Z.); (D.V.); (A.M.); (N.G.); (A.T.A.)
- Department of Dermatology and Venereology, Sestre milosrdnice University Hospital Centre, 10000 Zagreb, Croatia
| | - Petar Ozretić
- Laboratory for Hereditary Cancer, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia;
| | - Ivana Lojkić
- Croatian Veterinary Institute, 10000 Zagreb, Croatia;
| | - Marija Buljan
- School of Dental Medicine, University of Zagreb, 10000 Zagreb, Croatia; (L.L.M.); (M.B.); (M.Š.); (M.Z.); (D.V.); (A.M.); (N.G.); (A.T.A.)
- Department of Dermatology and Venereology, Sestre milosrdnice University Hospital Centre, 10000 Zagreb, Croatia
| | - Mirna Šitum
- School of Dental Medicine, University of Zagreb, 10000 Zagreb, Croatia; (L.L.M.); (M.B.); (M.Š.); (M.Z.); (D.V.); (A.M.); (N.G.); (A.T.A.)
- Department of Dermatology and Venereology, Sestre milosrdnice University Hospital Centre, 10000 Zagreb, Croatia
| | - Mario Zovak
- School of Dental Medicine, University of Zagreb, 10000 Zagreb, Croatia; (L.L.M.); (M.B.); (M.Š.); (M.Z.); (D.V.); (A.M.); (N.G.); (A.T.A.)
- Department of Surgery, Sestre milosrdnice University Hospital Centre, 10000 Zagreb, Croatia
| | - Dinko Vidović
- School of Dental Medicine, University of Zagreb, 10000 Zagreb, Croatia; (L.L.M.); (M.B.); (M.Š.); (M.Z.); (D.V.); (A.M.); (N.G.); (A.T.A.)
- Department of Surgery, Sestre milosrdnice University Hospital Centre, 10000 Zagreb, Croatia
| | - August Mijić
- School of Dental Medicine, University of Zagreb, 10000 Zagreb, Croatia; (L.L.M.); (M.B.); (M.Š.); (M.Z.); (D.V.); (A.M.); (N.G.); (A.T.A.)
- Department of Surgery, Sestre milosrdnice University Hospital Centre, 10000 Zagreb, Croatia
| | - Nada Galić
- School of Dental Medicine, University of Zagreb, 10000 Zagreb, Croatia; (L.L.M.); (M.B.); (M.Š.); (M.Z.); (D.V.); (A.M.); (N.G.); (A.T.A.)
| | - Arjana Tambić Andrašević
- School of Dental Medicine, University of Zagreb, 10000 Zagreb, Croatia; (L.L.M.); (M.B.); (M.Š.); (M.Z.); (D.V.); (A.M.); (N.G.); (A.T.A.)
- Department of Clinical Microbiology, University Hospital for Infectious Diseases, 10000 Zagreb, Croatia
| |
Collapse
|
14
|
Ćesić D, Lugović Mihić L, Ozretić P, Lojkić I, Buljan M, Šitum M, Zovak M, Vidović D, Mijić A, Galić N, Tambić Andrašević A. Association of Gut Lachnospiraceae and Chronic Spontaneous Urticaria. Life (Basel) 2023; 13:1280. [DOI: https:/doi.org/10.3390/life13061280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2023] Open
Abstract
(1) Background: Chronic spontaneous urticaria (CSU) has been linked to the dysbiosis of the gut microbiota. Furthermore, various studies have highlighted the anti-inflammatory properties of short-chain fatty acids (SCFAs), whose production is primarily regulated by the gut microbiota. However, only a few studies have investigated the role of major SCFA producers, such as Lachnospiraceae, in skin inflammatory diseases. (2) Goal: This study aimed to compare the abundance of Lachnospiraceae between CSU patients and healthy controls (HCs). (3) Material and methods: In this case–control study, 16S rRNA sequencing was performed to compare the composition of the gut microbiome between 22 CSU patients and 23 HCs. (4) Results: Beta-diversity revealed significant clustering (p < 0.05) between the CSU patients and HCs. Alpha diversity in the CSU group was significantly decreased according to the Evenness index (p < 0.05). The linear discriminant analysis effect size (LEfSe) identified the significant depletion of the Lachnospiraceae family in CSU patients. (5) Conclusion: Our study revealed the dysbiosis of the gut microbiota in CSU patients, including decreased levels of Lachnospiraceae members, responsible for SCFA production, suggesting that SCFAs may contribute to immune dysfunction in the pathogenesis of CSU. We speculate that the modulation of SCFAs could serve as a prospective additional option in CSU treatment.
Collapse
Affiliation(s)
- Diana Ćesić
- Department of Dermatology and Venereology, Medikol Clinic, 10000 Zagreb, Croatia
- School of Dental Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Liborija Lugović Mihić
- School of Dental Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Department of Dermatology and Venereology, Sestre milosrdnice University Hospital Centre, 10000 Zagreb, Croatia
| | - Petar Ozretić
- Laboratory for Hereditary Cancer, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Ivana Lojkić
- Croatian Veterinary Institute, 10000 Zagreb, Croatia
| | - Marija Buljan
- School of Dental Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Department of Dermatology and Venereology, Sestre milosrdnice University Hospital Centre, 10000 Zagreb, Croatia
| | - Mirna Šitum
- School of Dental Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Department of Dermatology and Venereology, Sestre milosrdnice University Hospital Centre, 10000 Zagreb, Croatia
| | - Mario Zovak
- School of Dental Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Department of Surgery, Sestre milosrdnice University Hospital Centre, 10000 Zagreb, Croatia
| | - Dinko Vidović
- School of Dental Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Department of Surgery, Sestre milosrdnice University Hospital Centre, 10000 Zagreb, Croatia
| | - August Mijić
- School of Dental Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Department of Surgery, Sestre milosrdnice University Hospital Centre, 10000 Zagreb, Croatia
| | - Nada Galić
- School of Dental Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Arjana Tambić Andrašević
- School of Dental Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Department of Clinical Microbiology, University Hospital for Infectious Diseases, 10000 Zagreb, Croatia
| |
Collapse
|
15
|
Roles of fermented plant-, dairy- and meat-based foods in the modulation of allergic responses. FOOD SCIENCE AND HUMAN WELLNESS 2023. [DOI: 10.1016/j.fshw.2022.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
16
|
Yamamoto-Hanada K, Sato M, Toyokuni K, Irahara M, Hiraide-Kotaki E, Harima-Mizusawa N, Morita H, Matsumoto K, Ohya Y. Combination of heat-killed Lactiplantibacillus plantarum YIT 0132 (LP0132) and oral immunotherapy in cow's milk allergy: a randomised controlled trial. Benef Microbes 2023; 14:17-30. [PMID: 36815492 DOI: 10.3920/bm2022.0064] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
Safer and more effective cow milk (CM)-oral immunotherapy that does not induce allergic reactions has not yet been standardised. We sought to explore the efficacy and feasibility of a combination of heat-killed Lactiplantibacillus plantarum YIT 0132 (LP0132) and oral immunotherapy for treating IgE-mediated cow milk allergy (CMA). We conducted a 24-week, double-blind, randomised (1:1), two-arm, parallel-group, placebo-controlled, phase 2 trial of LP0132 intervention for treating IgE-mediated CMA in children aged 1-18 years (n=60) from January 29, 2018 to July 12, 2019 in Tokyo, Japan. Participants were randomly assigned to the LP0132 group receiving citrus juice fermented with LP0132 or to the control group receiving citrus juice without. Both groups received low-dose slow oral immunotherapy with CM. The primary outcome was improved tolerance to CM, proven by the CM challenge test at 24 weeks. Secondary outcomes were changes in serum biomarkers of serum-specific β-lactoglobulin-IgE (sIgE) and β-lactoglobulin-IgG4 (sIgG4). Exploratory outcomes included changes in serum cytokine levels and gut microbiota composition. A total of 61 participants were included. Finally, 31 children were assigned to the LP0132 group and 30 to the control group, respectively. After the intervention, 41.4 and 37.9% of the participants in the LP0132 and control groups, respectively, showed improved tolerance to CM. In serum biomarkers after the intervention, the sIgG4 level was significantly higher, and interleukin (IL)-5 and IL-9 were significantly lower, in the LP0132 group than in the control group. In the gut microbiome, the α-diversity and Lachnospiraceae increased significantly in the LP0132 group, and Lachnospiraceae after the intervention was significantly higher in the LP0132 group than in the control group. In conclusion, low-dose oral immunotherapy with modulating gut microbiota might be a safer and more effective approach for treating cow's milk allergy.
Collapse
Affiliation(s)
- K Yamamoto-Hanada
- Allergy Center, National Center for Child Health and Development, 2-10-1 Okura, 1578535 Tokyo, Japan
| | - M Sato
- Allergy Center, National Center for Child Health and Development, 2-10-1 Okura, 1578535 Tokyo, Japan
| | - K Toyokuni
- Allergy Center, National Center for Child Health and Development, 2-10-1 Okura, 1578535 Tokyo, Japan
| | - M Irahara
- Allergy Center, National Center for Child Health and Development, 2-10-1 Okura, 1578535 Tokyo, Japan
| | - E Hiraide-Kotaki
- Yakult Central Institute for Microbiological Research, 5-11 Izumi, Kunitachi, Tokyo 186-8650, Japan
| | - N Harima-Mizusawa
- Yakult Central Institute for Microbiological Research, 5-11 Izumi, Kunitachi, Tokyo 186-8650, Japan
| | - H Morita
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, 2-10-1 Okura, 1578535 Tokyo, Japan
| | - K Matsumoto
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, 2-10-1 Okura, 1578535 Tokyo, Japan
| | - Y Ohya
- Allergy Center, National Center for Child Health and Development, 2-10-1 Okura, 1578535 Tokyo, Japan
| |
Collapse
|
17
|
Yu G, Zhang Q, Li H, Wang Y, Sheng H, Zhang S, Fu L. Effects of Allergen-Specific and Non-Specific AGEs on the Allergenicity of Ovalbumin in a Mouse Model of Food Allergy. Mol Nutr Food Res 2023; 67:e2200221. [PMID: 36550625 DOI: 10.1002/mnfr.202200221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
SCOPE Epidemiologic studies suggest a link between the incidence of food allergy and the consumption of dietary advanced glycation end-products (AGEs). However, the pathogenic role of dietary AGEs in food allergy is largely unknown. This study aims to investigate the effect of allergen-specific and non-specific AGEs on the allergenic manifestation of ovalbumin (OVA), a typical food allergen in vivo. METHODS AND RESULTS OVA is glycated by methylglyoxal to prepare allergen-specific AGEs (i.e., OVA-AGE), and a standard AIN-93G diet is heated to obtain allergen-non-specific AGEs. A BALB/c mouse model orally sensitizes to OVA with different forms of AGEs is established and the outcomes are measured as clinical signs, specific antibodies, type-2/type-2 cytokines, immune cell subpopulations, intestinal barrier function, and gut microbiota (GM) composition. The OVA-AGE which has a lower immunoglobulin E (IgE)-binding level in vitro does not reduce the allergenicity of OVA but promotes a stronger T helper 2 cells (Th2)-response than native OVA in vivo. Both forms of AGEs up-regulate the expression of splenic RAGE and aggravate the destruction of gut barrier and GM dysbiosis, especially when exposes to non-relevant AGEs. CONCLUSION This study highlights the role of dietary AGEs in food allergy and helps to understand the biological consequences of immune-toxic compounds in modern diet.
Collapse
Affiliation(s)
- Gang Yu
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, P. R. China
| | - Qiaozhi Zhang
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, P. R. China
| | - Huatao Li
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, P. R. China
| | - Yanbo Wang
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, P. R. China
| | - Huadong Sheng
- National Pre-packaged Food Quality Supervision and Inspection Center (Zhejiang), Zhejiang Fangyuan Test Group Co. LTD., Hangzhou, 310018, P. R. China
| | - Shuifeng Zhang
- National Pre-packaged Food Quality Supervision and Inspection Center (Zhejiang), Zhejiang Fangyuan Test Group Co. LTD., Hangzhou, 310018, P. R. China
| | - Linglin Fu
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, P. R. China
| |
Collapse
|
18
|
Huang YY, Liang YT, Wu JM, Wu WT, Liu XT, Ye TT, Chen XR, Zeng XA, Manzoor MF, Wang LH. Advances in the Study of Probiotics for Immunomodulation and Intervention in Food Allergy. Molecules 2023; 28:molecules28031242. [PMID: 36770908 PMCID: PMC9919562 DOI: 10.3390/molecules28031242] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/15/2023] [Accepted: 01/18/2023] [Indexed: 01/31/2023] Open
Abstract
Food allergies are a serious food safety and public health issue. Soybean, dairy, aquatic, poultry, and nut products are common allergens inducing allergic reactions and adverse symptoms such as atopic dermatitis, allergic eczema, allergic asthma, and allergic rhinitis. Probiotics are assumed as an essential ingredient in maintaining intestinal microorganisms' composition. They have unique physiological roles and therapeutic effects in maintaining the mucosal barrier, immune function, and gastrointestinal tract, inhibiting the invasion of pathogenic bacteria, and preventing diarrhea and food allergies. Multiple pieces of evidence reveal a significant disruptive effect of probiotics on food allergy pathology and progression mechanisms. Thus, this review describes the allergenic proteins as an entry point and briefly describes the application of probiotics in allergenic foods. Then, the role of probiotics in preventing and curing allergic diseases by regulating human immunity through intestinal flora and intestinal barrier, modulating host immune active cells, and improving host amino acid metabolism are described in detail. The anti-allergic role of probiotics in the function and metabolism of the gastrointestinal tract has been comprehensively explored to furnish insights for relieving food allergy symptoms and preventing food allergy.
Collapse
Affiliation(s)
- Yan-Yan Huang
- College of Food Science and Engineering, Foshan University, Foshan 528225, China
- Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing, Foshan University, Foshan 528225, China
| | - Yan-Tong Liang
- College of Food Science and Engineering, Foshan University, Foshan 528225, China
- Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing, Foshan University, Foshan 528225, China
| | - Jia-Min Wu
- College of Food Science and Engineering, Foshan University, Foshan 528225, China
- Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing, Foshan University, Foshan 528225, China
| | - Wei-Tong Wu
- College of Food Science and Engineering, Foshan University, Foshan 528225, China
- Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing, Foshan University, Foshan 528225, China
| | - Xin-Tong Liu
- College of Food Science and Engineering, Foshan University, Foshan 528225, China
- Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing, Foshan University, Foshan 528225, China
| | - Ting-Ting Ye
- College of Food Science and Engineering, Foshan University, Foshan 528225, China
- Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing, Foshan University, Foshan 528225, China
| | - Xiao-Rong Chen
- College of Food Science and Engineering, Foshan University, Foshan 528225, China
- Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing, Foshan University, Foshan 528225, China
| | - Xin-An Zeng
- College of Food Science and Engineering, Foshan University, Foshan 528225, China
- Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing, Foshan University, Foshan 528225, China
- Correspondence: (X.-A.Z.); (M.F.M.); (L.-H.W.)
| | - Muhammad Faisal Manzoor
- College of Food Science and Engineering, Foshan University, Foshan 528225, China
- Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing, Foshan University, Foshan 528225, China
- Correspondence: (X.-A.Z.); (M.F.M.); (L.-H.W.)
| | - Lang-Hong Wang
- College of Food Science and Engineering, Foshan University, Foshan 528225, China
- Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing, Foshan University, Foshan 528225, China
- Correspondence: (X.-A.Z.); (M.F.M.); (L.-H.W.)
| |
Collapse
|
19
|
Rousseaux A, Brosseau C, Bodinier M. Immunomodulation of B Lymphocytes by Prebiotics, Probiotics and Synbiotics: Application in Pathologies. Nutrients 2023; 15:nu15020269. [PMID: 36678140 PMCID: PMC9863037 DOI: 10.3390/nu15020269] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/22/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023] Open
Abstract
INTRODUCTION Prebiotics, probiotics and synbiotics are known to have major beneficial effects on human health due to their ability to modify the composition and the function of the gut mucosa, the gut microbiota and the immune system. These components largely function in a healthy population throughout different periods of life to confer homeostasis. Indeed, they can modulate the composition of the gut microbiota by increasing bacteria strands that are beneficial for health, such as Firmicute and Bifidobacteria, and decreasing harmful bacteria, such as Enteroccocus. Their immunomodulation properties have been extensively studied in different innate cells (dendritic cells, macrophages, monocytes) and adaptive cells (Th, Treg, B cells). They can confer a protolerogenic environment but also modulate pro-inflammatory responses. Due to all these beneficial effects, these compounds have been investigated to prevent or to treat different diseases, such as cancer, diabetes, allergies, autoimmune diseases, etc. Regarding the literature, the effects of these components on dendritic cells, monocytes and T cells have been studied and presented in a number of reviews, but their impact on B-cell response has been less widely discussed. CONCLUSIONS For the first time, we propose here a review of the literature on the immunomodulation of B-lymphocytes response by prebiotics, probiotics and synbiotics, both in healthy conditions and in pathologies. DISCUSSION Promising studies have been performed in animal models, highlighting the potential of prebiotics, probiotics and synbiotics intake to treat or to prevent diseases associated with B-cell immunomodulation, but this needs to be validated in humans with a full characterization of B-cell subsets and not only the humoral response.
Collapse
|
20
|
Moriki D, Francino MP, Koumpagioti D, Boutopoulou B, Rufián-Henares JÁ, Priftis KN, Douros K. The Role of the Gut Microbiome in Cow's Milk Allergy: A Clinical Approach. Nutrients 2022; 14:4537. [PMID: 36364799 PMCID: PMC9656688 DOI: 10.3390/nu14214537] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 07/30/2023] Open
Abstract
Cow's milk allergy (CMA) is the most prevalent food allergy (FA) in infancy and early childhood and can be present with various clinical phenotypes. The significant increase in FA rates recorded in recent decades has been associated with environmental and lifestyle changes that limit microbial exposure in early life and induce changes in gut microbiome composition. Gut microbiome is a diverse community of microbes that colonize the gastrointestinal tract (GIT) and perform beneficial functions for the host. This complex ecosystem interacts with the immune system and has a pivotal role in the development of oral tolerance to food antigens. Emerging evidence indicates that alterations of the gut microbiome (dysbiosis) in early life cause immune dysregulation and render the host susceptible to immune-mediated diseases later in life. Therefore, the colonization of the gut by "healthy" microbes that occurs in the first years of life determines the lifelong health of the host. Here, we present current data on the possible role of the gut microbiome in the development of CMA. Furthermore, we discuss how gut microbiome modification might be a potential strategy for CMA prevention and treatment.
Collapse
Affiliation(s)
- Dafni Moriki
- Allergology and Pulmonology Unit, 3rd Pediatric Department, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Maria Pilar Francino
- Department of Genomics and Health, Fundación Para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valencia (FISABIO), 46020 Valencia, Spain
- CIBER en Epidemiología y Salud Pública, 28001 Madrid, Spain
| | - Despoina Koumpagioti
- Department of Nursing, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Barbara Boutopoulou
- Department of Nursing, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - José Ángel Rufián-Henares
- Departamento de Nutrición y Bromatología, Instituto de Nutrición y Tecnología de los Alimentos, Centro de Investigación Biomédica, Universidad de Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria ibs. GRANADA, Universidad de Granada, 18071 Granada, Spain
| | - Kostas N. Priftis
- Allergology and Pulmonology Unit, 3rd Pediatric Department, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Konstantinos Douros
- Allergology and Pulmonology Unit, 3rd Pediatric Department, National and Kapodistrian University of Athens, 12462 Athens, Greece
| |
Collapse
|
21
|
Goh A, Muhardi L, Ali A, Liew WK, Estrada-Reyes E, Zepeda-Ortega B, Kudla U, van Neerven RJJ, Ulfman LH, Lambers TT, Warner JO. Differences between peptide profiles of extensive hydrolysates and their influence on functionality for the management of cow's milk allergy: A short review. FRONTIERS IN ALLERGY 2022; 3:950609. [PMID: 36660742 PMCID: PMC9843608 DOI: 10.3389/falgy.2022.950609] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 08/05/2022] [Indexed: 01/13/2023] Open
Abstract
Extensively hydrolyzed formulas (eHFs) are recommended for the dietary management of cow's milk protein allergy (CMPA) in non-exclusively breastfed infants. Studies show that peptide profiles differ between eHFs. This short review aims to highlight the variability in peptides and their ability to influence allergenicity and possibly the induction of tolerance by different eHFs. The differences between eHFs are determined by the source of the protein fraction (casein or whey), peptide size-distribution profile and residual β-lactoglobulin which is the most immunogenic and allergenic protein in bovine milk for human infants as it is not present in human breastmilk. These differences occur from the hydrolyzation process which result in variable IgE reactivity against cow's milk allergen epitopes by subjects with CMPA and differences in the Th1, Th2 and pro-inflammatory cytokine responses elicited. They also have different effects on gut barrier integrity. Results suggest that one particular eHF-casein had the least allergenic potential due to its low residual allergenic epitope content and demonstrated the greatest effect on restoring gut barrier integrity by its effects on mucin 5AC, occludin and Zona Occludens-1 in human enterocytes. It also increased the production of the tolerogenic cytokines Il-10 and IFN-γ. In addition, recent studies documented promising effects of optional functional ingredients such as pre-, pro- and synbiotics on the management of cow's milk allergy and induction of tolerance, in part via the induction of the production of short chain fatty acids. This review highlights differences in the residual allergenicity, peptide size distribution, presence of optional functional ingredients and overall functionality of several well-characterized eHFs which can impact the management of CMPA and the ability to induce immune tolerance to cow's milk protein.
Collapse
Affiliation(s)
- Anne Goh
- Department of Paediatrics, KK Women’s and Children’s Hospital, Singapore, Singapore,Correspondence: Anne Goh
| | - Leilani Muhardi
- Medical Affairs, Friesland Campina AMEA, Singapore, Singapore
| | - Adli Ali
- Department of Paediatrics, Universiti Kebangsaan Malaysia Medical Center, Bangi, Malaysia
| | - Woei Kang Liew
- Paediatric Allergy Immunology Rheumatology Centre, Mount Elizabeth Novena Specialist Centre, Singapore, Singapore
| | | | - Benjamin Zepeda-Ortega
- Department of Pediatrics, Angeles Lomas Hospital Huixquilucan Mexican State, Mexico, Mexico
| | | | - R. J. Joost van Neerven
- R&D, FrieslandCampina, Amersfoort, the Netherlands,Cell Biology and Immunology, Wageningen University, Wageningen, the Netherlands
| | | | | | - John O. Warner
- National Heart and Lung Institute, Imperial College, London, United Kingdom,Departement Pediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
22
|
Fiocchi A, Cabana MD, Mennini M. Current Use of Probiotics and Prebiotics in Allergy. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2022; 10:2219-2242. [PMID: 35792336 DOI: 10.1016/j.jaip.2022.06.038] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 06/27/2022] [Accepted: 06/27/2022] [Indexed: 01/06/2023]
Abstract
The microbiome plays an important role in the pathogenesis of allergic diseases. This review updates the reader on studies aimed at influencing allergic diseases through modulation of the gut microflora. A nonsystematic review of the literature was performed, focusing on relevant trials evaluating the effect of probiotics/prebiotics/symbiotics in the prevention and treatment of allergic disease. For each allergic disease, we were able to find not only a substantial number of clinical trials but also systematic reviews. Specific guidelines, based on systematic reviews and meta-analyses, are available for the prevention of allergic disease and for the treatment of food allergy. In each of the areas examined-allergic rhinitis, allergic asthma, atopic dermatitis, food allergy, and gastrointestinal allergies-there are substantial uncertainties in the efficacy of gut microflora modulation in prevention and treatment. At present, practicing clinicians can avail themselves of intestinal flora modulators as an adjunct in the prevention of atopic dermatitis but not of other forms of allergic diseases. Their effects on the treatment of allergic diseases remain controversial.
Collapse
Affiliation(s)
- Alessandro Fiocchi
- Translational Research in Pediatric Specialities Area, Allergy Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
| | - Michael D Cabana
- Department of Pediatrics, Albert Einstein College of Medicine and the Children's Hospital at Montefiore, Bronx, NY
| | - Maurizio Mennini
- Translational Research in Pediatric Specialities Area, Allergy Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
23
|
Augustine T, Kumar M, Al Khodor S, van Panhuys N. Microbial Dysbiosis Tunes the Immune Response Towards Allergic Disease Outcomes. Clin Rev Allergy Immunol 2022:10.1007/s12016-022-08939-9. [PMID: 35648372 DOI: 10.1007/s12016-022-08939-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2022] [Indexed: 02/07/2023]
Abstract
The hygiene hypothesis has been popularized as an explanation for the rapid increase in allergic disease observed over the past 50 years. Subsequent epidemiological studies have described the protective effects that in utero and early life exposures to an environment high in microbial diversity have in conferring protective benefits against the development of allergic diseases. The rapid advancement in next generation sequencing technology has allowed for analysis of the diverse nature of microbial communities present in the barrier organs and a determination of their role in the induction of allergic disease. Here, we discuss the recent literature describing how colonization of barrier organs during early life by the microbiota influences the development of the adaptive immune system. In parallel, mechanistic studies have delivered insight into the pathogenesis of disease, by demonstrating the comparative effects of protective T regulatory (Treg) cells, with inflammatory T helper 2 (Th2) cells in the development of immune tolerance or induction of an allergic response. More recently, a significant advancement in our understanding into how interactions between the adaptive immune system and microbially derived factors play a central role in the development of allergic disease has emerged. Providing a deeper understanding of the symbiotic relationship between our microbiome and immune system, which explains key observations made by the hygiene hypothesis. By studying how perturbations that drive dysbiosis of the microbiome can cause allergic disease, we stand to benefit by delineating the protective versus pathogenic aspects of human interactions with our microbial companions, allowing us to better harness the use of microbial agents in the design of novel prophylactic and therapeutic strategies.
Collapse
Affiliation(s)
- Tracy Augustine
- Laboratory of Immunoregulation, Sidra Medicine, PO BOX 26999, Doha, Qatar
| | - Manoj Kumar
- Microbiome and Host-Microbes Interactions Laboratory, Sidra Medicine, Doha, Qatar
| | - Souhaila Al Khodor
- Microbiome and Host-Microbes Interactions Laboratory, Sidra Medicine, Doha, Qatar
| | | |
Collapse
|
24
|
Contribution of Gut Microbiota to Immune Tolerance in Infants. J Immunol Res 2022; 2021:7823316. [PMID: 34993254 PMCID: PMC8727111 DOI: 10.1155/2021/7823316] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 11/18/2021] [Indexed: 02/07/2023] Open
Abstract
The prevalence of food allergy has increased in recent years, especially among the pediatric population. Differences in the gut microbiota composition between children with FA and healthy children have brought this topic into the spotlight as a possible explanation for the increase in FA. The gut microbiota characteristics are acquired through environmental interactions starting early in life, such as type of delivery during birth and breastfeeding. The microbiota features may be shaped by a plethora of immunomodulatory mechanisms, including a predominant role of Tregs and the transcription factor FOXP3. Additionally, a pivotal role has been given to vitamin A and butyrate, the main anti-inflammatory metabolite. These observations have led to the study and development of therapies oriented to modifying the microbiota and metabolite profiles, such as the use of pre- and probiotics and the determination of their capacity to induce tolerance to allergens that are relevant to FA. To date, evidence supporting these approaches in humans is scarce and inconclusive. Larger cohorts and dose-titration studies are mandatory to evaluate whether the observed changes in gut microbiota composition reflect medical recovery and increased tolerance in pediatric patients with FA. In this article, we discuss the establishment of the microbiota, the immunological mechanisms that regulate the microbiota of children with food allergies, and the evidence in research focused on its regulation as a means to achieve tolerance to food allergens.
Collapse
|
25
|
Ashraf SA, Elkhalifa AEO, Ahmad MF, Patel M, Adnan M, Sulieman AME. Probiotic Fermented Foods and Health Promotion. AFRICAN FERMENTED FOOD PRODUCTS- NEW TRENDS 2022:59-88. [DOI: 10.1007/978-3-030-82902-5_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
26
|
Wang X, Yi W, He L, Luo S, Wang J, Jiang L, Long H, Zhao M, Lu Q. Abnormalities in Gut Microbiota and Metabolism in Patients With Chronic Spontaneous Urticaria. Front Immunol 2021; 12:691304. [PMID: 34721374 PMCID: PMC8554312 DOI: 10.3389/fimmu.2021.691304] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 09/29/2021] [Indexed: 01/09/2023] Open
Abstract
Background Increasing evidence suggests that the gut microbiome plays a role in the pathogenesis of allergy and autoimmunity. The association between abnormalities in the gut microbiota and chronic spontaneous urticaria (CSU) remains largely undefined. Methods Fecal samples were obtained from 39 patients with CSU and 40 healthy controls (HCs). 16S ribosomal RNA (rRNA) gene sequencing (39 patients with CSU and 40 HCs) and untargeted metabolomics (12 patients with CSU and 12 HCs) were performed to analyze the compositional and metabolic alterations of the gut microbiome in CSU patients and HCs. Results The 16S rRNA gene sequencing results showed a significant difference in the β-diversity of the gut microbiota, presented as the Jaccard distance, between CSU patients and HCs. No significant differences were found in the α-diversity of the gut microbiota between patients and HCs. At the phylum level, the major bacteria in the gut microbiome of patients with CSU were Firmicutes, Bacteroidetes, Proteobacteria, and Actinobacteria. At the genus level, Lactobacillus, Turicibacter, and Lachnobacterium were significantly increased and Phascolarctobacterium was decreased in patients with CSU. PICRUSt and correlation analysis indicated that Lactobacillus, Turicibacter, and Phascolarctobacterium were positively related to G protein-coupled receptors. Metabolomic analysis showed that α-mangostin and glycyrrhizic acid were upregulated and that 3-indolepropionic acid, xanthine, and isobutyric acid were downregulated in patients with CSU. Correlation analysis between the intestinal microbiota and metabolites suggested that there was a positive correlation between Lachnobacterium and α-mangostin. Conclusions This study suggests that disturbances in the gut microbiome composition and metabolites and their crosstalk or interaction may participate in the pathogenesis of CSU.
Collapse
Affiliation(s)
- Xin Wang
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Wanyu Yi
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Liting He
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shuaihantian Luo
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jiaqi Wang
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Li Jiang
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Hai Long
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ming Zhao
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Qianjin Lu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China.,Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| |
Collapse
|
27
|
Yang Y, Li X, Yang Y, Shoaie S, Zhang C, Ji B, Wei Y. Advances in the Relationships Between Cow's Milk Protein Allergy and Gut Microbiota in Infants. Front Microbiol 2021; 12:716667. [PMID: 34484158 PMCID: PMC8415629 DOI: 10.3389/fmicb.2021.716667] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 07/22/2021] [Indexed: 12/24/2022] Open
Abstract
Cow's milk protein allergy (CMPA) is an immune response to cow's milk proteins, which is one of the most common food allergies in infants and young children. It is estimated that 2-3% of infants and young children have CMPA. The diet, gut microbiota, and their interactions are believed to be involved in the alterations of mucosal immune tolerance, which might lead to the development of CMPA and other food allergies. In this review, the potential molecular mechanisms of CMPA, including omics technologies used for analyzing microbiota, impacts of early microbial exposures on CMPA development, and microbiota-host interactions, are summarized. The probiotics, prebiotics, synbiotics, fecal microbiota transplantation, and other modulation strategies for gut microbiota and the potential application of microbiota-based design of diets for the CMPA treatment are also discussed. This review not only summarizes the current studies about the interactions of CMPA with gut microbiota but also gives insights into the possible CMPA treatment strategies by modulating gut microbiota, which might help in improving the life quality of CMPA patients in the future.
Collapse
Affiliation(s)
- Yudie Yang
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Xiaoqi Li
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Ying Yang
- Jing’an District Central Hospital of Shanghai, Jing’an Branch, Huashan Hospital, Fudan University, Shanghai, China
| | - Saeed Shoaie
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
- Faculty of Dentistry, Oral and Craniofacial Sciences, Centre for Host-Microbiome Interactions, King’s College London, London, United Kingdom
| | - Cheng Zhang
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Boyang Ji
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Yongjun Wei
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Laboratory of Synthetic Biology, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
28
|
D’Auria E, Salvatore S, Acunzo M, Peroni D, Pendezza E, Di Profio E, Fiore G, Zuccotti GV, Verduci E. Hydrolysed Formulas in the Management of Cow's Milk Allergy: New Insights, Pitfalls and Tips. Nutrients 2021; 13:2762. [PMID: 34444922 PMCID: PMC8401609 DOI: 10.3390/nu13082762] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/03/2021] [Accepted: 08/07/2021] [Indexed: 12/12/2022] Open
Abstract
An allergy to cow's milk requires the avoidance of cow's milk proteins and, in some infants, the use of a hypoallergenic formula. This review aims to summarize the current evidence concerning different types of hydrolysed formulas (HF), and recommendations for the treatment of IgE- and non-IgE-mediated cow's milk allergy and functional gastrointestinal disorders in infancy, for which some dietary intervention and HF may be of benefit to both immune and motor mechanisms. Current guidelines recommend cow's milk protein (i.e., whey or casein) extensively hydrolysed formula (eHF) as the first choice for cow's milk allergy treatment, and amino acid formulas for more severe cases or those with reactions to eHF. Rice hydrolysed formulas (rHF) have also become available in recent years. Both eHF and rHF are well tolerated by the majority of children allergic to cow's milk, with no concerns regarding body growth or adverse effects. Some hydrolysates may have a pro-active effect in modulating the immune system due to the presence of small peptides and additional components, like biotics. Despite encouraging results on tolerance acquisition, evidence is still not conclusive, thus hampering our ability to draw firm conclusions. In clinical practice, the choice of hypoallergenic formula should be based on the infant's age, the severity, frequency and persistence of symptoms, immune phenotype, growth pattern, formula cost, and in vivo proof of tolerance and efficacy.
Collapse
Affiliation(s)
- Enza D’Auria
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (M.A.); (E.P.); (E.D.P.); (G.F.); (G.V.Z.); (E.V.)
| | - Silvia Salvatore
- Department of Pediatrics, Ospedale “F. Del Ponte”, University of Insubria, 21100 Varese, Italy;
| | - Miriam Acunzo
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (M.A.); (E.P.); (E.D.P.); (G.F.); (G.V.Z.); (E.V.)
| | - Diego Peroni
- Department of Clinical and Experimental Medicine, Section of Pediatrics, University of Pisa, 56126 Pisa, Italy;
| | - Erica Pendezza
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (M.A.); (E.P.); (E.D.P.); (G.F.); (G.V.Z.); (E.V.)
| | - Elisabetta Di Profio
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (M.A.); (E.P.); (E.D.P.); (G.F.); (G.V.Z.); (E.V.)
| | - Giulia Fiore
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (M.A.); (E.P.); (E.D.P.); (G.F.); (G.V.Z.); (E.V.)
| | - Gian Vincenzo Zuccotti
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (M.A.); (E.P.); (E.D.P.); (G.F.); (G.V.Z.); (E.V.)
| | - Elvira Verduci
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (M.A.); (E.P.); (E.D.P.); (G.F.); (G.V.Z.); (E.V.)
| |
Collapse
|
29
|
Chiu CJ, Huang MT. Asthma in the Precision Medicine Era: Biologics and Probiotics. Int J Mol Sci 2021; 22:4528. [PMID: 33926084 PMCID: PMC8123613 DOI: 10.3390/ijms22094528] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/23/2021] [Accepted: 04/24/2021] [Indexed: 02/07/2023] Open
Abstract
Asthma is a major global health issue. Over 300 million people worldwide suffer from this chronic inflammatory airway disease. Typical clinical symptoms of asthma are characterized by a recurrent wheezy cough, chest tightness, and shortness of breath. The main goals of asthma management are to alleviate asthma symptoms, reduce the risk of asthma exacerbations, and minimize long-term medicinal adverse effects. However, currently available type 2 T helper cells (Th2)-directed treatments are often ineffective due to the heterogeneity of the asthma subgroups, which manifests clinically with variable and poor treatment responses. Personalized precision therapy of asthma according to individualized clinical characteristics (phenotype) and laboratory biomarkers (endotype) is the future prospect. This mini review discusses the molecular mechanisms underlying asthma pathogenesis, including the hot sought-after topic of microbiota, add-on therapies and the potential application of probiotics in the management of asthma.
Collapse
Affiliation(s)
- Chiao-Juno Chiu
- Graduate Institute of Clinical Medicine, School of Medicine, National Taiwan University, Taipei 100, Taiwan;
| | - Miao-Tzu Huang
- Graduate Institute of Clinical Medicine, School of Medicine, National Taiwan University, Taipei 100, Taiwan;
- Department of Medical Research, National Taiwan University Hospital, No. 7 Chung-Shan South Road, Taipei 100, Taiwan
- Department of Pediatrics, National Taiwan University Hospital, No. 7 Chung-Shan South Road, Taipei 100, Taiwan
| |
Collapse
|
30
|
Murdaca G, Gerosa A, Paladin F, Petrocchi L, Banchero S, Gangemi S. Vitamin D and Microbiota: Is There a Link with Allergies? Int J Mol Sci 2021; 22:4288. [PMID: 33924232 PMCID: PMC8074777 DOI: 10.3390/ijms22084288] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/11/2021] [Accepted: 04/19/2021] [Indexed: 12/12/2022] Open
Abstract
There is increasing recognition of the importance of both the microbiome and vitamin D in states of health and disease. Microbiome studies have already demonstrated unique microbial patterns in systemic autoimmune diseases such as inflammatory bowel disease, rheumatoid arthritis, and systemic lupus erythematosus. Dysbiosis also seems to be associated with allergies, in particular asthma, atopic dermatitis, and food allergy. Even though the effect of vitamin D supplementation on these pathologies is still unknown, vitamin D deficiency deeply influences the microbiome by altering the microbiome composition and the integrity of the gut epithelial barrier. It also influences the immune system mainly through the vitamin D receptor (VDR). In this review, we summarize the influence of the microbiome and vitamin D on the immune system with a particular focus on allergic diseases and we discuss the necessity of further studies on the use of probiotics and of a correct intake of vitamin D.
Collapse
Affiliation(s)
- Giuseppe Murdaca
- Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (A.G.); (F.P.); (L.P.); (S.B.)
- Ospedale Policlinico San Martino IRCCS, 16132 Genoa, Italy
| | - Alessandra Gerosa
- Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (A.G.); (F.P.); (L.P.); (S.B.)
- Ospedale Policlinico San Martino IRCCS, 16132 Genoa, Italy
| | - Francesca Paladin
- Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (A.G.); (F.P.); (L.P.); (S.B.)
- Ospedale Policlinico San Martino IRCCS, 16132 Genoa, Italy
| | - Lorena Petrocchi
- Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (A.G.); (F.P.); (L.P.); (S.B.)
- Ospedale Policlinico San Martino IRCCS, 16132 Genoa, Italy
| | - Sara Banchero
- Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (A.G.); (F.P.); (L.P.); (S.B.)
- Ospedale Policlinico San Martino IRCCS, 16132 Genoa, 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
|
31
|
Albuhairi S, Rachid R. Biologics and Novel Therapies for Food Allergy. Immunol Allergy Clin North Am 2021; 41:271-283. [PMID: 33863483 DOI: 10.1016/j.iac.2021.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Food allergy is a significant public health burden affecting around 10% of adults and 8% of children. Although the first peanut oral immunotherapy product received Food and Drug Administration approval in 2020, there is still an unmet need for more effective therapeutic options that minimize the risk of anaphylaxis, nutritional deficiencies, and patient's quality of life. Biologics are promising modalities, as they may improve compliance, target multiple food allergies, and treat other concomitant atopic diseases. Although omalizumab has been evaluated extensively, most biologics are more novel and have broader immunologic impact. Careful evaluation of their safety profile should therefore be conducted.
Collapse
Affiliation(s)
- Sultan Albuhairi
- Department of Pediatrics, Allergy and Immunology Section, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Rima Rachid
- Division of Immunology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
32
|
Lopez-Santamarina A, Gonzalez EG, Lamas A, Mondragon ADC, Regal P, Miranda JM. Probiotics as a Possible Strategy for the Prevention and Treatment of Allergies. A Narrative Review. Foods 2021; 10:foods10040701. [PMID: 33806092 PMCID: PMC8064452 DOI: 10.3390/foods10040701] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 02/06/2023] Open
Abstract
Allergies are an increasing global public health concern, especially for children and people living in urban environments. Allergies impair the quality of life of those who suffer from them, and for this reason, alternatives for the treatment of allergic diseases or reduction in their symptoms are being sought. The main objective of this study was to compile the studies carried out on probiotics as a possible therapy for allergies. The most studied allergies on which probiotics have been shown to have a beneficial effect are rhinitis, asthma, and atopic dermatitis. Most studies have studied the administration of Lactobacillus and Bifidobacterium spp. in children and have shown beneficial effects, such as a reduction in hyperreactivity and inflammation caused by allergens and a decrease in cytokine release, among other beneficial effects. In the case of children, no clear beneficial effects were found in several studies, and the potential risk from the use of some opportunistic bacteria, such as probiotics, seems controversial. In the studies that reported beneficial results, these effects were found to make allergy symptoms less aggressive, thus reducing morbidity in allergy sufferers. The different effects of the same probiotic bacteria on different patients seem to reinforce the idea that the efficacy of probiotics is dependent on the microbial species or strain, its derived metabolites and byproducts, and the gut microbiota eubiosis of the patient. This study is relevant in the context of allergic diseases, as it provides a broader understanding of new alternatives for the treatment of allergies, both in children, who are the main sufferers, and adults, showing that probiotics, in some cases, reduce the symptoms and severity of such diseases.
Collapse
|
33
|
Larnkjær A, Michaelsen KF, Rytter MJH, Mølgaard C, Laursen RP. Effect of probiotics on thymus size and markers of infection in late infancy: a randomized controlled trial. Pediatr Res 2021; 89:563-568. [PMID: 32305995 DOI: 10.1038/s41390-020-0895-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/24/2020] [Accepted: 03/30/2020] [Indexed: 11/09/2022]
Abstract
BACKGROUND Probiotics are known to stimulate the immune system but the effect on thymus size in late infancy is unknown. We examined the effect of probiotics on thymus size and C-reactive protein (CRP) in healthy Danish infants starting daycare. We further examined associations between thymus size, CRP and recent infections. METHODS The study included 186 children randomized to a combination of Lactobacillus rhamnosus, LGG® and Bifidobacterium animalis spp. lactis, BB-12® or placebo for 6 months. Thymus size, assessed as thymus index (TI) and thymus weight index (TWI), was measured by ultrasound at baseline and at endpoint. Blood samples were drawn to measure CRP. Infections were parent-reported. RESULTS There was no significant difference in thymus size between the probiotic group and placebo (p ≥ 0.248) but TWI tended to be higher in the probiotic group corresponding to 5% higher than placebo (p = 0.068) in an adjusted model. There was no effect of probiotics on CRP (p = 0.331). At the endpoint, thymus size was inversely associated with CRP (p ≤ 0.040), diarrhea (p ≤ 0.050), and TI was also associated with the absence from daycare due to respiratory or gastrointestinal infections (p = 0.010). CONCLUSION The probiotic intervention had no effect on thymus size or CRP in Danish children at the age of starting daycare. IMPACT Overall there was no effect on thymus size of a combination of Lactobacillus rhamnosus, LGG® and Bifidobacterium animalis spp. lactis, BB-12® administered to Danish children starting daycare. This study examines the effect of probiotics on thymus size in healthy children when they start daycare thus exposed for infections while their immune system is still developing. This has to our knowledge not been described before. We found no significant difference in thymus size between the probiotic and placebo groups, but for thymus weight index, there was a trend. This should be investigated further in studies designed for this as primary outcome.
Collapse
Affiliation(s)
- Anni Larnkjær
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark.
| | - Kim F Michaelsen
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Maren J H Rytter
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark.,Department of Pediatrics, Slagelse Hospital, Slagelse, Denmark
| | - Christian Mølgaard
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Rikke P Laursen
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
34
|
Suther C, Moore MD, Beigelman A, Zhou Y. The Gut Microbiome and the Big Eight. Nutrients 2020; 12:nu12123728. [PMID: 33287179 PMCID: PMC7761723 DOI: 10.3390/nu12123728] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/28/2020] [Accepted: 12/01/2020] [Indexed: 12/11/2022] Open
Abstract
Food allergies are increasing at an alarming rate, with 6.5% of the general population affected. It has been hypothesized that the increase in allergies stems from the “hygiene hypothesis”. The gut microbiome, a collection of microbiota and their genetic contents from the gastrointestinal tract, has been shown to play a part in the development of food allergies. The Food and Drug Administration requires all regulated food companies to clearly state an inclusion of the major, or “big eight” food allergens on packaging. This review is to provide information on the significant advancements related to the gut microbiome and each of the eight major food allergies individually. Establishment of causal connection between the microbiome and food allergies has uncovered novel mechanisms. New strategies are discussed to prevent future sensitization and reaction through novel treatments involving functional additives and dietary changes that target the microbiome.
Collapse
Affiliation(s)
- Cassandra Suther
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA; (C.S.); (M.D.M.)
- Department of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Matthew D. Moore
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA; (C.S.); (M.D.M.)
| | - Avraham Beigelman
- Kipper Institute of Allergy and Immunology, Schneider Children’s Medical Center, Tel Aviv University, Tel Aviv 5891000, Israel;
| | - Yanjiao Zhou
- Department of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA
- Correspondence: ; Tel.: +1-860-679-6379
| |
Collapse
|
35
|
Łoś-Rycharska E, Gołębiewski M, Grzybowski T, Rogalla-Ładniak U, Krogulska A. The microbiome and its impact on food allergy and atopic dermatitis in children. Postepy Dermatol Alergol 2020; 37:641-650. [PMID: 33240001 PMCID: PMC7675070 DOI: 10.5114/ada.2019.90120] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 08/29/2019] [Indexed: 01/23/2023] Open
Abstract
Food allergy (FA) affects 4-10% of children, especially children with atopic dermatitis (AD). During infancy the gut microbiome may determine both the course of FA and tolerance to food allergens. Analogically, the skin microbiome changes in the course of AD. Most studies have associated FA with a lower abundance and diversity of Lactobacillales and Clostridiales, but greater numbers of Enterobacterales, while AD in children has been associated with lower numbers of Staphylococcus epidermidis and S. hominis but an abundance of S. aureus and Streptococcus species. An understanding of the impact of the microbiome on the clinical course of FA and AD may allow for the development of new models of allergy treatment and prevention.
Collapse
Affiliation(s)
- Ewa Łoś-Rycharska
- Department of Paediatrics, Allergology and Gastroenterology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Marcin Gołębiewski
- Interdisciplinary Centre of Modern Technologies, Nicolaus Copernicus University, Torun, Poland
- Chair of Plant Physiology and Biotechnology, Nicolaus Copernicus University, Torun, Poland
| | - Tomasz Grzybowski
- Chair of Forensic Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Urszula Rogalla-Ładniak
- Chair of Forensic Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Aneta Krogulska
- Department of Paediatrics, Allergology and Gastroenterology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| |
Collapse
|
36
|
Abstract
Food allergens are innocuous proteins that promote tolerogenic adaptive immune responses in healthy individuals yet in other individuals induce an allergic adaptive immune response characterized by the presence of antigen-specific immunoglobulin E and type-2 immune cells. The cellular and molecular processes that determine a tolerogenic versus non-tolerogenic immune response to dietary antigens are not fully elucidated. Recently, there have been advances in the identification of roles for microbial communities and anatomical sites of dietary antigen exposure and presentation that have provided new insights into the key regulatory steps in the tolerogenic versus non-tolerogenic decision-making processes. Herein, we will review and discuss recent findings in cellular and molecular processes underlying food sensitization and tolerance, immunological processes underlying severity of food-induced anaphylaxis, and insights obtained from immunotherapy trials.
Collapse
Affiliation(s)
- Sunil Tomar
- 1. Mary H. Weiser Food Allergy Center, Department of Pathology, University of Michigan 4051-BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Simon P Hogan
- 1. Mary H. Weiser Food Allergy Center, Department of Pathology, University of Michigan 4051-BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| |
Collapse
|
37
|
Navarro-Tapia E, Sebastiani G, Sailer S, Toledano LA, Serra-Delgado M, García-Algar Ó, Andreu-Fernández V. Probiotic Supplementation During the Perinatal and Infant Period: Effects on Gut Dysbiosis and Disease. Nutrients 2020; 12:E2243. [PMID: 32727119 PMCID: PMC7468726 DOI: 10.3390/nu12082243] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/15/2020] [Accepted: 07/22/2020] [Indexed: 02/07/2023] Open
Abstract
The perinatal period is crucial to the establishment of lifelong gut microbiota. The abundance and composition of microbiota can be altered by several factors such as preterm delivery, formula feeding, infections, antibiotic treatment, and lifestyle during pregnancy. Gut dysbiosis affects the development of innate and adaptive immune responses and resistance to pathogens, promoting atopic diseases, food sensitization, and infections such as necrotizing enterocolitis (NEC). Recent studies have indicated that the gut microbiota imbalance can be restored after a single or multi-strain probiotic supplementation, especially mixtures of Lactobacillus and Bifidobacterium strains. Following the systematic search methodology, the current review addresses the importance of probiotics as a preventive or therapeutic tool for dysbiosis produced during the perinatal and infant period. We also discuss the safety of the use of probiotics in pregnant women, preterm neonates, or infants for the treatment of atopic diseases and infections.
Collapse
Affiliation(s)
- Elisabet Navarro-Tapia
- Grup de Recerca Infancia i Entorn (GRIE), Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Valencian International University (VIU), 46002 Valencia, Spain
| | - Giorgia Sebastiani
- Department of Neonatology, Hospital Clínic-Maternitat, ICGON, BCNatal, 08028 Barcelona, Spain
| | - Sebastian Sailer
- Department of Neonatology, Hospital Clínic-Maternitat, ICGON, BCNatal, 08028 Barcelona, Spain
| | - Laura Almeida Toledano
- Institut de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
- BCNatal, Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), University of Barcelona, 08950 Barcelona, Spain
| | - Mariona Serra-Delgado
- Institut de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
- BCNatal, Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), University of Barcelona, 08950 Barcelona, Spain
| | - Óscar García-Algar
- Grup de Recerca Infancia i Entorn (GRIE), Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Department of Neonatology, Hospital Clínic-Maternitat, ICGON, BCNatal, 08028 Barcelona, Spain
| | - Vicente Andreu-Fernández
- Grup de Recerca Infancia i Entorn (GRIE), Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Valencian International University (VIU), 46002 Valencia, Spain
- Department of Neonatology, Hospital Clínic-Maternitat, ICGON, BCNatal, 08028 Barcelona, Spain
| |
Collapse
|
38
|
Stróżyk A, Horvath A, Meyer R, Szajewska H. Efficacy and safety of hydrolyzed formulas for cow's milk allergy management: A systematic review of randomized controlled trials. Clin Exp Allergy 2020; 50:766-779. [PMID: 32446273 DOI: 10.1111/cea.13669] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 05/12/2020] [Accepted: 05/17/2020] [Indexed: 12/26/2022]
Abstract
OBJECTIVE To summarize evidence on the efficacy and safety of the use of extensively hydrolyzed formulas (EHFs) for the treatment of children with cow's milk allergy (CMA). DESIGN Systematic review of randomized controlled trials (RCTs) per PRISMA guidelines. The risk of bias of included RCTs was assessed using the Cochrane Collaboration's risk of bias tool. In general, a narrative synthesis of the findings was performed. When sufficient data were available, a meta-analysis using the random-effect model was performed. DATA SOURCES The Cochrane Library, MEDLINE, and EMBASE databases were searched up to February 2020. ELIGIBILITY CRITERIA RCTs, including cross-over trials, assessing children of any age with any type of CMA that compared use of a formula containing extensively hydrolyzed bovine proteins (whey and/or casein) with use of any other formula for CMA management, were eligible for inclusion. Each type of EHF was evaluated separately. Outcome measures included allergic reactions (ie gastrointestinal, dermatological, and respiratory symptoms), growth, tolerance acquisition to cow's milk proteins, health-related quality of life, and safety. RESULTS Fifteen trials reported in 18 publications (1285 children) fulfilled the inclusion criteria. The study findings were limited by numerous methodological issues, including differences in outcome measures and their definitions, lack of pre-specified protocols and/or trial registration, and poor reporting of adverse events, methods of sequence generation and allocation concealment. The EHF products evaluated to date appear to be well-tolerated by most children with CMA. However, published studies do not allow for any conclusion to be reached regarding the benefit of one formula over another formula intended for CMA management. CONCLUSIONS This systematic review highlights the need for standardized treatment protocols, including an agreed-upon standardized set of outcomes that should be measured and reported in all clinical trials of specialized milk formula for the management of CMA.
Collapse
Affiliation(s)
- Agata Stróżyk
- Department of Paediatrics, The Medical University of Warsaw, Warsaw, Poland
| | - Andrea Horvath
- Department of Paediatrics, The Medical University of Warsaw, Warsaw, Poland
| | | | - Hania Szajewska
- Department of Paediatrics, The Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
39
|
Nance CL, Deniskin R, Diaz VC, Paul M, Anvari S, Anagnostou A. The Role of the Microbiome in Food Allergy: A Review. CHILDREN-BASEL 2020; 7:children7060050. [PMID: 32466620 PMCID: PMC7346163 DOI: 10.3390/children7060050] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/14/2020] [Accepted: 05/17/2020] [Indexed: 02/06/2023]
Abstract
Food allergies are common and estimated to affect 8% of children and 11% of adults in the United States. They pose a significant burden—physical, economic and social—to those affected. There is currently no available cure for food allergies. Emerging evidence suggests that the microbiome contributes to the development and manifestations of atopic disease. According to the hygiene hypothesis, children growing up with older siblings have a lower incidence of allergic disease compared with children from smaller families, due to their early exposure to microbes in the home. Research has also demonstrated that certain environmental exposures, such as a farming environment, during early life are associated with a diverse bacterial experience and reduced risk of allergic sensitization. Dysregulation in the homeostatic interaction between the host and the microbiome or gut dysbiosis appears to precede the development of food allergy, and the timing of such dysbiosis is critical. The microbiome affects food tolerance via the secretion of microbial metabolites (e.g., short chain fatty acids) and the expression of microbial cellular components. Understanding the biology of the microbiome and how it interacts with the host to maintain gut homeostasis is helpful in developing smarter therapeutic approaches. There are ongoing trials evaluating the benefits of probiotics and prebiotics, for the prevention and treatment of atopic diseases to correct the dysbiosis. However, the routine use of probiotics as an intervention for preventing allergic disease is not currently recommended. A new approach in microbial intervention is to attempt a more general modification of the gut microbiome, such as with fecal microbiota transplantation. Developing targeted bacterial therapies for food allergy may be promising for both the treatment and prevention of food allergy. Similarly, fecal microbiota transplantation is being explored as a potentially beneficial interventional approach. Overall, targeted bacterial therapies for food allergy may be promising for both the treatment and prevention of food allergy.
Collapse
Affiliation(s)
- Christina L. Nance
- Baylor College of Medicine, Section of Pediatric Immunology, Allergy and Retrovirology, Houston, TX 77030 USA; (C.L.N.); (R.D.); (V.C.D.); (M.P.); (S.A.)
- Texas Children’s Hospital, Department of Pediatrics, Section of Immunology, Allergy and Retrovirology, Houston, TX 77030, USA
| | - Roman Deniskin
- Baylor College of Medicine, Section of Pediatric Immunology, Allergy and Retrovirology, Houston, TX 77030 USA; (C.L.N.); (R.D.); (V.C.D.); (M.P.); (S.A.)
- Texas Children’s Hospital, Department of Pediatrics, Section of Immunology, Allergy and Retrovirology, Houston, TX 77030, USA
| | - Veronica C. Diaz
- Baylor College of Medicine, Section of Pediatric Immunology, Allergy and Retrovirology, Houston, TX 77030 USA; (C.L.N.); (R.D.); (V.C.D.); (M.P.); (S.A.)
- Texas Children’s Hospital, Department of Pediatrics, Section of Immunology, Allergy and Retrovirology, Houston, TX 77030, USA
| | - Misu Paul
- Baylor College of Medicine, Section of Pediatric Immunology, Allergy and Retrovirology, Houston, TX 77030 USA; (C.L.N.); (R.D.); (V.C.D.); (M.P.); (S.A.)
- Texas Children’s Hospital, Department of Pediatrics, Section of Immunology, Allergy and Retrovirology, Houston, TX 77030, USA
| | - Sara Anvari
- Baylor College of Medicine, Section of Pediatric Immunology, Allergy and Retrovirology, Houston, TX 77030 USA; (C.L.N.); (R.D.); (V.C.D.); (M.P.); (S.A.)
- Texas Children’s Hospital, Department of Pediatrics, Section of Immunology, Allergy and Retrovirology, Houston, TX 77030, USA
| | - Aikaterini Anagnostou
- Baylor College of Medicine, Section of Pediatric Immunology, Allergy and Retrovirology, Houston, TX 77030 USA; (C.L.N.); (R.D.); (V.C.D.); (M.P.); (S.A.)
- Texas Children’s Hospital, Department of Pediatrics, Section of Immunology, Allergy and Retrovirology, Houston, TX 77030, USA
- Correspondence:
| |
Collapse
|
40
|
Peroni DG, Nuzzi G, Trambusti I, Di Cicco ME, Comberiati P. Microbiome Composition and Its Impact on the Development of Allergic Diseases. Front Immunol 2020; 11:700. [PMID: 32391012 PMCID: PMC7191078 DOI: 10.3389/fimmu.2020.00700] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/27/2020] [Indexed: 12/24/2022] Open
Abstract
Allergic diseases, such as food allergy (FA), atopic dermatitis (AD), and asthma, are heterogeneous inflammatory immune-mediated disorders that currently constitute a public health issue in many developed countries worldwide. The significant increase in the prevalence of allergic diseases reported over the last few years has closely paralleled substantial environmental changes both on a macro and micro scale, which have led to reduced microbial exposure in early life and perturbation of the human microbiome composition. Increasing evidence shows that early life interactions between the human microbiome and the immune cells play a pivotal role in the development of the immune system. Therefore, the process of early colonization by a “healthy” microbiome is emerging as a key determinant of life-long health. In stark contrast, the perturbation of such a process, which results in changes in the host-microbiome biodiversity and metabolic activities, has been associated with greater susceptibility to immune-mediated disorders later in life, including allergic diseases. Here, we outline recent findings on the potential contribution of the microbiome in the gastrointestinal tract, skin, and airways to the development of FA, AD, and asthma. Furthermore, we address how the modulation of the microbiome composition in these different body districts could be a potential strategy for the prevention and treatment of allergic diseases.
Collapse
Affiliation(s)
- Diego G Peroni
- Department of Clinical and Experimental Medicine, Section of Pediatrics, University of Pisa, Pisa, Italy
| | - Giulia Nuzzi
- Department of Clinical and Experimental Medicine, Section of Pediatrics, University of Pisa, Pisa, Italy
| | - Irene Trambusti
- Department of Clinical and Experimental Medicine, Section of Pediatrics, University of Pisa, Pisa, Italy
| | - Maria Elisa Di Cicco
- Department of Clinical and Experimental Medicine, Section of Pediatrics, University of Pisa, Pisa, Italy
| | - Pasquale Comberiati
- Department of Clinical and Experimental Medicine, Section of Pediatrics, University of Pisa, Pisa, Italy.,Department of Clinical Immunology and Allergology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
41
|
|
42
|
Santos SCD, Konstantyner T, Cocco RR. Effects of probiotics in the treatment of food hypersensitivity in children: a systematic review. Allergol Immunopathol (Madr) 2020; 48:95-104. [PMID: 31477401 DOI: 10.1016/j.aller.2019.04.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 04/25/2019] [Accepted: 04/29/2019] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Food allergy is considered a public health problem for children. The modulation of the intestinal microbiota seems a promising strategy for the control of allergic reactions. OBJECTIVE To describe the effects of different forms of probiotics in pediatric food hypersensitivity treatment. DATA SOURCE We conducted a systematic review based on clinical trials published in the PubMed and Web of Science databases. The searches were carried out using the MeSH terms "Food Hypersensitivity," "Probiotics," "Lactobacillus," and "Bifidobacterium". DATA SYNTHESIS The final selection resulted in 18 clinical trials, which were predominantly samples of infants and pre-school children. The most-often used strain, either alone or in combination, was Lactobacillus rhamnosus GG; a placebo was mainly used in the control group. As for the vehicle, the most common forms were capsules and infant formulas, and the period of intervention ranged from four weeks to 24 months, with weekly or monthly visits to measure the outcomes. In these 18 trials, 46 analyses were performed with 27 different types of outcomes to evaluate the effects of probiotics (12 laboratory and 15 clinical). Twenty-seven of these analyses demonstrated the benefits of using these microorganisms. The SCORAD (atopic dermatitis index) and IgE levels and cytokines were the outcomes mostly evaluated. CONCLUSION The use of probiotics is beneficial in promoting immunomodulation and reducing clinical symptoms. However, more methodologically based research is needed to clarify the effect from each type, dose, and time of using them for the establishment of definitive care protocols.
Collapse
|
43
|
Kim BG, Kim JN, Jang AS, Shin M. Combined Effects of Lactobacillus rhamnosus and Egg Oral Immunotherapy in a Mouse Model of Egg Allergy. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2020; 12:701-711. [PMID: 32400134 PMCID: PMC7225004 DOI: 10.4168/aair.2020.12.4.701] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 01/17/2020] [Accepted: 02/04/2020] [Indexed: 01/22/2023]
Abstract
Purpose Recent clinical trials have successfully used oral immunotherapy (OIT) to treat food allergies. Probiotics have immunomodulatory effects by balancing Th1/Th2 immunity and enhancing regulatory T-cell activity. In this study, we analyzed the effects of OIT, probiotics alone, and probiotics administered simultaneously with OIT in a mouse model of egg allergy. Methods C3H/HeJ mice were sensitized by intragastric administration of ovomucoid (OM) with cholera toxin. For the OIT regime, increasing doses of OM were administered orally to sensitized mice. Lactobacillus casei variety ramnosus (Lcr35) was also administered. The mice were divided into 4 groups: control (no OIT), OIT, Lcr35, and OIT plus Lcr35 (OIT + Lcr35). The effects of OIT and Lcr35 treatment were estimated based on the symptom score, rectal temperature, serum levels of OM-specific immunoglobulin (Ig)E, IgA, IgG1, and IgG2a immediately after and 2 weeks after ceasing treatment and histological staining of the small intestine. Results The severity of anaphylaxis decreased in all treatment groups. Simultaneous administration of Lcr35 and OIT decreased the severity of anaphylaxis compared to controls and the OIT group. The protective effects were sustained 2 weeks after ceasing treatment in all treatment groups. A significant decrease in OM-specific IgA, IgG1, and IgG2a levels was observed in both the OIT and OIT plus Lcr35 groups. However, a significant decrease in the OM-specific IgE level was observed only in OIT plus Lcr35 treated mice and was sustained 2 weeks after ceasing treatment. Mucin amounts in the small intestine decreased after OIT, OIT plus Lcr35, and Lcr35 treatment with the lowest in the OIT plus Lcr35 group. Conclusions Lcr35 treatment during OIT had some synergic effect for protection against anaphylaxis in a mice model of egg allergy. These findings should be confirmed in future animal studies including more detailed immunological profiles and human studies.
Collapse
Affiliation(s)
- Byeong Gon Kim
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University School of Medicine, Bucheon, Korea
| | - Ji Na Kim
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University School of Medicine, Bucheon, Korea
| | - An Soo Jang
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University School of Medicine, Bucheon, Korea
| | - Meeyong Shin
- Department of Pediatrics, Soonchunhyang University Bucheon Hospital, Soonchunhyang University School of Medicine, Bucheon, Korea.
| |
Collapse
|
44
|
Sestito S, D'Auria E, Baldassarre ME, Salvatore S, Tallarico V, Stefanelli E, Tarsitano F, Concolino D, Pensabene L. The Role of Prebiotics and Probiotics in Prevention of Allergic Diseases in Infants. Front Pediatr 2020; 8:583946. [PMID: 33415087 PMCID: PMC7783417 DOI: 10.3389/fped.2020.583946] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 11/23/2020] [Indexed: 12/15/2022] Open
Abstract
Allergic diseases have been linked to genetic and/or environmental factors, such as antibiotic use, westernized high fat and low fiber diet, which lead to early intestinal dysbiosis, and account for the rise in allergy prevalence, especially in western countries. Allergic diseases have shown reduced microbial diversity, including fewer lactobacilli and bifidobacteria, within the neonatal microbiota, before the onset of atopic diseases. Raised interest in microbiota manipulating strategies to restore the microbial balance for atopic disease prevention, through prebiotics, probiotics, or synbiotics supplementation, has been reported. We reviewed and discussed the role of prebiotics and/or probiotics supplementation for allergy prevention in infants. We searched PubMed and the Cochrane Database using keywords relating to "allergy" OR "allergic disorders," "prevention" AND "prebiotics" OR "probiotics" OR "synbiotics." We limited our evaluation to papers of English language including children aged 0-2 years old. Different products or strains used, different period of intervention, duration of supplementation, has hampered the draw of definitive conclusions on the clinical impact of probiotics and/or prebiotics for prevention of allergic diseases in infants, except for atopic dermatitis in infants at high-risk. This preventive effect on eczema in high-risk infants is supported by clear evidence for probiotics but only moderate evidence for prebiotic supplementation. However, the optimal prebiotic or strain of probiotic, dose, duration, and timing of intervention remains uncertain. Particularly, a combined pre- and post-natal intervention appeared of stronger benefit, although the definition of the optimal intervention starting time during gestation, the timing, and duration in the post-natal period, as well as the best target population, are still an unmet need.
Collapse
Affiliation(s)
- Simona Sestito
- Pediatric Unit, Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Enza D'Auria
- Department of Pediatrics, Vittore Buzzi Children's Hospital-University of Milan, Milan, Italy
| | - Maria Elisabetta Baldassarre
- Neonatology and Neonatal Intensive Care Unit, Department of Biomedical Science and Human Oncology, "Aldo Moro" University of Bari, Bari, Italy
| | - Silvia Salvatore
- Department of Pediatrics, Ospedale "F. Del Ponte", University of Insubria, Varese, Italy
| | - Valeria Tallarico
- Pediatric Unit, Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Ettore Stefanelli
- Pediatric Unit, Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Flora Tarsitano
- Pediatric Unit, Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Daniela Concolino
- Pediatric Unit, Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.,Department of Health Sciences, School of Medicine and Surgery, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Licia Pensabene
- Pediatric Unit, Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| |
Collapse
|
45
|
Bunyavanich S, Berin MC. Food allergy and the microbiome: Current understandings and future directions. J Allergy Clin Immunol 2019; 144:1468-1477. [PMID: 31812181 PMCID: PMC6905201 DOI: 10.1016/j.jaci.2019.10.019] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/21/2019] [Accepted: 10/23/2019] [Indexed: 02/07/2023]
Abstract
Growing evidence points to an important role for the commensal microbiota in susceptibility to food allergy. Epidemiologic studies demonstrate associations between exposures known to modify the microbiome and risk of food allergy. Direct profiling of the gut microbiome in human cohort studies has demonstrated that individuals with food allergy have distinct gut microbiomes compared to healthy control subjects, and dysbiosis precedes the development of food allergy. Mechanistic studies in mouse models of food allergy have confirmed that the composition of the intestinal microbiota can imprint susceptibility or resistance to food allergy on the host and have identified a unique population of microbially responsive RORγt-positive FOXp3-positive regulatory T cells as critical for the maintenance of tolerance to foods. Armed with this new understanding of the role of the microbiota in food allergy and tolerance, therapeutics aimed at modifying the gastrointestinal microbiota are in development. In this article we review key milestones in the development of our current understanding of how the gastrointestinal microbiota contributes to food allergy and discuss our vision for the future of the field.
Collapse
Affiliation(s)
- Supinda Bunyavanich
- Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY.
| | - M Cecilia Berin
- Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY; PRIISM Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
46
|
Qamer S, Deshmukh M, Patole S. Probiotics for cow's milk protein allergy: a systematic review of randomized controlled trials. Eur J Pediatr 2019; 178:1139-1149. [PMID: 31230196 DOI: 10.1007/s00431-019-03397-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 05/01/2019] [Accepted: 05/06/2019] [Indexed: 12/26/2022]
Abstract
Cow's milk protein allergy (CMPA) is the commonest food allergy in infancy and is associated with significant health burden. Given their immune modulatory properties, probiotics have been proposed as a strategy for management of CMPA. We aimed to systematically review efficacy and safety of probiotics in the management of CMPA. Databases PubMed, EMBASE, CINAHL, Cochrane Central Library, and Google scholar were searched in August 2018 for randomized controlled trials (RCT) of probiotic supplementation as an adjunct in the management of infants with suspected/proven CMPA. Primary outcomes were resolution of hematochezia and acquisition of tolerance to CMP at 6, 12, 24, and 36 months. Secondary outcomes included effect on allergic symptoms (SCORAD index), growth, gut microbiota, and adverse effects. A total of 10 RCTs (n = 845; probiotics, 422; control, 423) with low to unclear risk of bias were included. Meta-analysis showed probiotic supplementation was not associated with earlier resolution of hematochezia (n = 87; RR: 1.45 (95% CI: 0.96-2.18), p = 0.08; level of evidence (LOE), very low), in presumed CMPA. In confirmed CMPA, probiotics were associated with higher rate of acquisition of tolerance to CMP at the end of 3 years compared with placebo (N = 493; RR, 1.47; 95% CI, (1.17-1.84); p = 0.0009; LOE, low]. Meta-analysis was not possible for other outcomes. There were no probiotic related adverse effects. Conclusion: Limited low-quality evidence indicates that probiotic supplementation may be associated with earlier acquisition of tolerance to CMP in children with CMPA. Large well-designed trials are essential to confirm these findings. What is Known: • Cow's milk protein allergy (CMPA) is one of the commonest food allergies in children. CMPA is associated with significant socioeconomic burden. • Elimination diet and extensively hydrolyzed formula is the mainstay of the management of CMPA. What is New: • This first systematic review of randomized controlled trials shows that probiotics as an adjuvant can lead to earlier acquisition of tolerance to CMP in children at 36 months of age. However, the evidence is low quality and influenced by data from one large study. • Probiotic supplementation was not associated with earlier resolution of hematochezia.
Collapse
Affiliation(s)
- Sheeba Qamer
- Department of Neonatal Paediatrics, King Edward Memorial Hospital, Perth, WA, Australia
| | - Mangesh Deshmukh
- Department of Neonatalogy, Fiona Stanley Hospital, 11 Robin warren drive, Perth, WA, 6150, Australia.
- Department of Neonatalogy, St. John of God Hospital, Subiaco, Perth, WA, 6008, Australia.
| | - Sanjay Patole
- Department of Neonatal Paediatrics, King Edward Memorial Hospital, Perth, WA, Australia
| |
Collapse
|
47
|
D'Auria E, Salvatore S, Pozzi E, Mantegazza C, Sartorio MUA, Pensabene L, Baldassarre ME, Agosti M, Vandenplas Y, Zuccotti G. Cow's Milk Allergy: Immunomodulation by Dietary Intervention. Nutrients 2019; 11:1399. [PMID: 31234330 PMCID: PMC6627562 DOI: 10.3390/nu11061399] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 06/14/2019] [Accepted: 06/17/2019] [Indexed: 12/26/2022] Open
Abstract
Cow's milk proteins cause allergic symptoms in 2% to 3% of all infants. In these individuals, the physiological mechanism of tolerance is broken with subsequent possible sensitization to antigens, which can lead eventually to allergic responses. The present review aims to provide an overview of different aspects of immune modulation by dietary intervention in cow's milk allergy (CMA). It focuses on pathogenetic mechanisms of different CMA related disorders, e.g., gastroesophageal reflux and eosinophilic esophagitis, highlighting the role of dietary management on innate and adaptive immune systems. The traditional dietary management of CMA has greatly changed in the last years, moving from a passive approach, consisting of an elimination diet to relieve symptoms, to a "proactive" one, meaning the possibility to actively modulate the immune system. Thus, new insights into the role of hydrolysates and baked milk in immunomodulation are addressed here. Additionally, nutritional components, such as pre- and probiotics, may target the immune system via microbiota, offering a possible road map for new CMA prevention and treatment strategies.
Collapse
Affiliation(s)
- Enza D'Auria
- Department of Pediatrics, Vittore Buzzi Children's Hospital-University of Milan, 20154 Milan, Italy.
| | - Silvia Salvatore
- Department of Pediatrics, Ospedale "F. Del Ponte", University of Insubria, 21100 Varese, Italy.
| | - Elena Pozzi
- Department of Pediatrics, Vittore Buzzi Children's Hospital-University of Milan, 20154 Milan, Italy.
| | - Cecilia Mantegazza
- Department of Pediatrics, Vittore Buzzi Children's Hospital-University of Milan, 20154 Milan, Italy.
| | - Marco Ugo Andrea Sartorio
- Department of Pediatrics, Vittore Buzzi Children's Hospital-University of Milan, 20154 Milan, Italy.
| | - Licia Pensabene
- Department of Medical and Surgical Sciences, Pediatric Unit, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy.
| | - Maria Elisabetta Baldassarre
- Neonatology and Neonatal Intensive Care Unit, Department of Biomedical Science and Human Oncology, "Aldo Moro" University of Bari, P.zza Giulio Cesare 11, 70124 Bari, Italy.
| | - Massimo Agosti
- Department of Pediatrics, Ospedale "F. Del Ponte", University of Insubria, 21100 Varese, Italy.
| | - Yvan Vandenplas
- KidZ Health Castle, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium.
| | - GianVincenzo Zuccotti
- Department of Pediatrics, Vittore Buzzi Children's Hospital-University of Milan, 20154 Milan, Italy.
| |
Collapse
|
48
|
Licari A, Manti S, Marseglia A, Brambilla I, Votto M, Castagnoli R, Leonardi S, Marseglia GL. Food Allergies: Current and Future Treatments. MEDICINA (KAUNAS, LITHUANIA) 2019; 55:120. [PMID: 31052434 PMCID: PMC6571952 DOI: 10.3390/medicina55050120] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 04/27/2019] [Accepted: 04/27/2019] [Indexed: 12/22/2022]
Abstract
Food allergies are an increasingly public health problem, affecting up to 10% of children and causing a significant burden on affected patients, resulting in dietary restrictions, fear of accidental ingestion and related risk of severe reactions, as well as a reduced quality of life. Currently, there is no specific cure for a food allergy, so the only available management is limited to strict dietary avoidance, education on prompt recognition of symptoms, and emergency treatment of adverse reactions. Several allergen specific- and nonspecific-therapies, aiming to acquire a persistent food tolerance, are under investigation as potential treatments; however, to date, only immunotherapy has been identified as the most promising therapeutic approach for food allergy treatment. The aim of this review is to provide an updated overview on changes in the treatment landscape for food allergies.
Collapse
Affiliation(s)
- Amelia Licari
- Pediatric Clinic, Department of Pediatrics, Fondazione IRCCS Policlinico San Matteo, University of Pavia, 27100 Pavia, Italy.
| | - Sara Manti
- Department of Pediatrics, Unit of Pediatric Genetics and Immunology, University of Messina, 98122 Messina, Italy.
- Department of Clinical and Experimental Medicine, University of Catania, 95131 Catania, Italy.
| | | | - Ilaria Brambilla
- Pediatric Clinic, Department of Pediatrics, Fondazione IRCCS Policlinico San Matteo, University of Pavia, 27100 Pavia, Italy.
| | - Martina Votto
- Pediatric Clinic, Department of Pediatrics, Fondazione IRCCS Policlinico San Matteo, University of Pavia, 27100 Pavia, Italy.
| | - Riccardo Castagnoli
- Pediatric Clinic, Department of Pediatrics, Fondazione IRCCS Policlinico San Matteo, University of Pavia, 27100 Pavia, Italy.
| | - Salvatore Leonardi
- Department of Clinical and Experimental Medicine, University of Catania, 95131 Catania, Italy.
| | - Gian Luigi Marseglia
- Pediatric Clinic, Department of Pediatrics, Fondazione IRCCS Policlinico San Matteo, University of Pavia, 27100 Pavia, Italy.
| |
Collapse
|
49
|
Zhao W, Ho HE, Bunyavanich S. The gut microbiome in food allergy. Ann Allergy Asthma Immunol 2019; 122:276-282. [PMID: 30578857 PMCID: PMC6389411 DOI: 10.1016/j.anai.2018.12.012] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 12/13/2018] [Accepted: 12/13/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To review observational human, murine, and interventional trial studies that have examined the gut microbiome in food allergy, and to provide perspective on future investigations in this field. DATA SOURCES A review of the published literature was performed with PubMed, and clinical studies catalogued at ClinicalTrials.gov were also reviewed. STUDY SELECTIONS The most recent relevant studies, seminal works, and topical clinical trials were selected. RESULTS Gut dysbiosis likely precedes the development of food allergy, and the timing of such dysbiosis is critical. Gut microbiota associated with individual food allergies may be distinct. Murine models support the importance of gut microbiota in shaping immune maturation and tolerance. Gut microbiota may affect food allergy susceptibility by modulating type 2 immunity, influencing immune development and tolerance, regulating basophil populations, and promoting intestinal barrier function. Ongoing and future interventional trials of probiotics, prebiotics, synbiotics, and fecal microbiota transfer will help translate our understanding of the gut microbiome in food allergy to clinical practice. Future work in this area will include deepening of current research foci, as well as expansion of efforts to include the virome, mycobiome, and interactions between the microbiome, host, and environment. Robust and consistent study designs, multidimensional profiling, and systems biology approaches will enable this future work. CONCLUSION By advancing research on the microbiome in food allergy, we can further our understanding of food allergy and derive new approaches for its prevention and therapy.
Collapse
Affiliation(s)
- William Zhao
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Hsi-En Ho
- Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Supinda Bunyavanich
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
50
|
Berni Canani R, Paparo L, Nocerino R, Di Scala C, Della Gatta G, Maddalena Y, Buono A, Bruno C, Voto L, Ercolini D. Gut Microbiome as Target for Innovative Strategies Against Food Allergy. Front Immunol 2019; 10:191. [PMID: 30828329 PMCID: PMC6384262 DOI: 10.3389/fimmu.2019.00191] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 01/22/2019] [Indexed: 12/11/2022] Open
Abstract
The dramatic increase in food allergy prevalence and severity globally requires effective strategies. Food allergy derives from a defect in immune tolerance mechanisms. Immune tolerance is modulated by gut microbiota function and structure, and microbiome alterations (dysbiosis) have a pivotal role in the development of food allergy. Environmental factors, including a low-fiber/high-fat diet, cesarean delivery, antiseptic agents, lack of breastfeeding, and drugs can induce gut microbiome dysbiosis, and have been associated with food allergy. New experimental tools and technologies have provided information regarding the role of metabolites generated from dietary nutrients and selected probiotic strains that could act on immune tolerance mechanisms. The mechanisms are multiple and still not completely defined. Increasing evidence has provided useful information on optimal bacterial species/strains, dosage, and timing for intervention. The increased knowledge of the crucial role played by nutrients and gut microbiota-derived metabolites is opening the way to a post-biotic approach in the stimulation of immune tolerance through epigenetic regulation. This review focused on the potential role of gut microbiome as the target for innovative strategies against food allergy.
Collapse
Affiliation(s)
- Roberto Berni Canani
- Department of Translational Medical Science – Pediatric Section, University of Naples “Federico II”, Naples, Italy
- European Laboratory for the Investigation of Food-Induced Diseases, University of Naples “Federico II”, Naples, Italy
- ImmunoNutritionLab at CEINGE-Advanced Biotechnologies, University of Naples “Federico II”, Naples, Italy
- Task Force on Microbiome Studies, University of Naples “Federico II”, Naples, Italy
| | - Lorella Paparo
- Department of Translational Medical Science – Pediatric Section, University of Naples “Federico II”, Naples, Italy
- ImmunoNutritionLab at CEINGE-Advanced Biotechnologies, University of Naples “Federico II”, Naples, Italy
| | - Rita Nocerino
- Department of Translational Medical Science – Pediatric Section, University of Naples “Federico II”, Naples, Italy
- ImmunoNutritionLab at CEINGE-Advanced Biotechnologies, University of Naples “Federico II”, Naples, Italy
| | - Carmen Di Scala
- Department of Translational Medical Science – Pediatric Section, University of Naples “Federico II”, Naples, Italy
- ImmunoNutritionLab at CEINGE-Advanced Biotechnologies, University of Naples “Federico II”, Naples, Italy
| | - Giusy Della Gatta
- Department of Translational Medical Science – Pediatric Section, University of Naples “Federico II”, Naples, Italy
- ImmunoNutritionLab at CEINGE-Advanced Biotechnologies, University of Naples “Federico II”, Naples, Italy
| | - Ylenia Maddalena
- Department of Translational Medical Science – Pediatric Section, University of Naples “Federico II”, Naples, Italy
| | - Aniello Buono
- Department of Translational Medical Science – Pediatric Section, University of Naples “Federico II”, Naples, Italy
- ImmunoNutritionLab at CEINGE-Advanced Biotechnologies, University of Naples “Federico II”, Naples, Italy
| | - Cristina Bruno
- Department of Translational Medical Science – Pediatric Section, University of Naples “Federico II”, Naples, Italy
- ImmunoNutritionLab at CEINGE-Advanced Biotechnologies, University of Naples “Federico II”, Naples, Italy
| | - Luana Voto
- Department of Translational Medical Science – Pediatric Section, University of Naples “Federico II”, Naples, Italy
| | - Danilo Ercolini
- Task Force on Microbiome Studies, University of Naples “Federico II”, Naples, Italy
- Department of Agricultural Sciences, University of Naples “Federico II”, Naples, Italy
| |
Collapse
|