1
|
Sarrabayrouse G, Joulain C, Bessoles S, Chiron AS, Abina AM, Hacein-Bey-Abina S. Erythropoietin supplementation induces dysbiosis of the gut microbiota and impacts mucosal immunity in a non-diseased mouse model. Front Immunol 2025; 15:1465410. [PMID: 39916952 PMCID: PMC11798978 DOI: 10.3389/fimmu.2024.1465410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 12/20/2024] [Indexed: 02/09/2025] Open
Abstract
A number of drug treatments are known to alter the dialogue between the gut microbiota and the immune system components in the digestive mucosa. Alterations in intestinal homeostasis are now well known to affect peripheral immune responses and favor the occurrence of a number of pathologies such as allergies and cancers. Erythropoietin's known pleiotropic effects might explain the adverse events sometimes observed in anemic patients treated by erythropoiesis-stimulating agents (ESA). However, the impact of this therapeutic cytokine on the homeostasis of the intestinal tract has not previously been investigated in detail. By studying a mouse model of erythropoietin (EPO) supplementation for 28 days, we observed EPO-induced dysbiosis of the fecal microbiota characterized by a greater bacterial load, lower bacterial diversity and taxonomic changes. With regard to the mucosal immune system, an analysis of leukocyte populations in the small intestine and colon treatment revealed low proportions of ileal CD4 lymphocyte subpopulations (Treg, Tr17 and Th17 cells), IgA-secreting plasma cells, and a major macrophage subpopulation, involved in the control of lymphocyte responses. Our results provide for the first time a descriptive analysis of intestinal EPO's regulatory properties and raise questions about the involvement of EPO-induced alterations in the microbiota and the gut immune effectors in the control of intestinal and peripheral immune responses.
Collapse
Affiliation(s)
- Guillaume Sarrabayrouse
- Unité des technologies Chimiques et Biologiques pour la Santé, Université Paris Cité, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), UTCBS, Paris, France
| | - Corentin Joulain
- Unité des technologies Chimiques et Biologiques pour la Santé, Université Paris Cité, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), UTCBS, Paris, France
| | - Stéphanie Bessoles
- Unité des technologies Chimiques et Biologiques pour la Santé, Université Paris Cité, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), UTCBS, Paris, France
| | - Andrada S. Chiron
- Unité des technologies Chimiques et Biologiques pour la Santé, Université Paris Cité, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), UTCBS, Paris, France
- Clinical Immunology Laboratory, Groupe Hospitalier Universitaire Paris-Saclay, Hôpital Bicêtre, Assistance Publique-Hôpitaux de Paris, Le-Kremlin-Bicêtre, France
| | - Amine M. Abina
- Unité des technologies Chimiques et Biologiques pour la Santé, Université Paris Cité, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), UTCBS, Paris, France
| | - Salima Hacein-Bey-Abina
- Unité des technologies Chimiques et Biologiques pour la Santé, Université Paris Cité, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), UTCBS, Paris, France
- Clinical Immunology Laboratory, Groupe Hospitalier Universitaire Paris-Saclay, Hôpital Bicêtre, Assistance Publique-Hôpitaux de Paris, Le-Kremlin-Bicêtre, France
| |
Collapse
|
2
|
Malekiantaghi A, Ghanaati F, Shabani-Mirzaee H, Shariat M, Mojtahedi SY, Eftekhari K. Lactobacillus rhamnosus Helps to Reduce the Duration of Bleeding in Breastfed Infants with Allergic Proctocolitis. Breastfeed Med 2025; 20:59-64. [PMID: 39417285 DOI: 10.1089/bfm.2024.0185] [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] [Indexed: 10/19/2024]
Abstract
Background: Cow's milk protein allergy (CMPA) is the most common food allergy in infants. The current standard of care (SOC) involves eliminating the allergen from both the infant's and mother's diet for 2-4 weeks. The purpose of this study is to assess the effectiveness of Lactobacillus rhamnosus (Ramnoflor) in reducing the duration of bleeding in these infants. Methods: This randomized clinical trial was conducted at Bahrami Children's Hospital on breastfed infants who were diagnosed with CMPA and had a positive occult blood (OB) test. Patients were randomly assigned to either the control or case groups. All patients received SOC therapy, with the case group receiving Ramnoflor and the control group receiving a placebo. Data were recorded on the checklist, and the children were followed and visited three times during the study, with an OB assessment at each visit. Results: The study enrolled 48 infants. Among the infants in the case group, the OB test was positive in four cases (8.3%) on the fifth day. However, there were no positive cases on the 14th and 30th days. The prevalence of this test was significantly lower in patients who received probiotics compared to the control group on the fifth day (p < 0.001). There were no positive OB tests on the 14th and 30th days in any of the groups, and no significant difference was observed between the groups. Conclusion: The addition of L. rhamnosus to SOC therapy led to a decrease in the duration of rectal bleeding in infants with CMPA compared to the control group.
Collapse
Affiliation(s)
- Armen Malekiantaghi
- Department of Pediatric, Bahrami Children's Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Faezeh Ghanaati
- Department of Pediatric, Bahrami Children's Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hosein Shabani-Mirzaee
- Department of Pediatric Endocrinology, Bahrami Children's Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mamak Shariat
- Maternal, Fetal & Neonatal Research Center, Family Health Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sayed-Yousef Mojtahedi
- Department of Pediatric Nephrology, Bahrami Children's Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kambiz Eftekhari
- Department of Pediatric, Pediatric Gastroenterology and Hepatology Research Center, Bahrami Children's Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Vallianatou GN, Douladiris N, Mageiros L, Manousakis E, Zisaki V, Galani M, Xepapadaki P, Taka S, Papadopoulos NG. Duration of food protein-induced allergic proctocolitis (FPIAP) and the role of intestinal microbiota. Pediatr Allergy Immunol 2024; 35:e70008. [PMID: 39629903 PMCID: PMC11616471 DOI: 10.1111/pai.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 10/31/2024] [Accepted: 11/14/2024] [Indexed: 12/08/2024]
Abstract
BACKGROUND Food protein-induced allergic proctocolitis (FPIAP) is the leading cause of rectal bleeding in infants. Tolerance is presumed to develop until the first year of age, although natural history studies are scarce, making the determination of the ideal duration for any intervention, challenging. Intestinal microbiota (IM) is crucial in food allergy development; however, data for FPIAP remain limited. This study aimed to assess FPIAP remission after 3 months of milk avoidance and its correlation with IM longitudinal changes. METHODS A prospective observational study of infants aged ≤6 months with a diagnosis of FPIAP. After 3 months of management according to a clinical algorithm, infants were subjected to a milk challenge using either a cow (CM) or a goat (GM) milk formula in a random order. Stool samples were collected longitudinally for microbiome analysis. RESULTS Out of 61 infants, 57 were challenged (29 with CM, 28 with GM). Of these, 55 (96.5%) achieved tolerance, with no difference in tolerance rates between CM (28/29) and GM (27/28). The average age of tolerance development was 6.3 months. Enterobacteriaceae clusters (Klebsiella- and Shigella-dominated) were most often represented in samples from symptomatic infants. In contrast, Bacteroides and Bifidobacteria clusters emerged later, in apparently healthy infants. CONCLUSION A 3-month intervention was sufficient for almost all infants to achieve tolerance. GM was tolerated equally well to CM. Symptomatic FPIAP is associated with immature enterotypes, while disease remission coincides with microbiome changes in time.
Collapse
Affiliation(s)
- G. N. Vallianatou
- Allergy Department, 2nd Pediatric ClinicNational and Kapodistrian University of AthensAthensGreece
| | - N. Douladiris
- Allergy Department, 2nd Pediatric ClinicNational and Kapodistrian University of AthensAthensGreece
| | - L. Mageiros
- Allergy Department, 2nd Pediatric ClinicNational and Kapodistrian University of AthensAthensGreece
- Department of Information Technology and Biomedical SciencesThe American College of GreeceAthensGreece
| | - E. Manousakis
- Allergy Department, 2nd Pediatric ClinicNational and Kapodistrian University of AthensAthensGreece
| | - V. Zisaki
- Allergy Department, 2nd Pediatric ClinicNational and Kapodistrian University of AthensAthensGreece
| | - M. Galani
- Allergy Department, 2nd Pediatric ClinicNational and Kapodistrian University of AthensAthensGreece
| | - P. Xepapadaki
- Allergy Department, 2nd Pediatric ClinicNational and Kapodistrian University of AthensAthensGreece
| | - S. Taka
- Allergy Department, 2nd Pediatric ClinicNational and Kapodistrian University of AthensAthensGreece
- Startbio PC Molecular Diagnostics and Biotechnology ServicesStartbioAthensGreece
| | - N. G. Papadopoulos
- Allergy Department, 2nd Pediatric ClinicNational and Kapodistrian University of AthensAthensGreece
- University of ManchesterManchesterUK
| |
Collapse
|
4
|
Augustine T, Murugesan S, Badri F, Gentilcore G, Grivel JC, Akobeng A, Elawad M, Adeli M, Al Khodor S, van Panhuys N. Immunoglobulin-coating patterns reveal altered humoral responses to gut bacteria in pediatric cow milk allergies. J Transl Med 2024; 22:1021. [PMID: 39533360 PMCID: PMC11558889 DOI: 10.1186/s12967-024-05850-z] [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: 08/05/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Pediatric cow milk allergies (CMA) can occur in immunoglobulin (Ig) E and non-IgE-mediated forms. Unlike IgE-mediated allergies, the mechanisms of disease pathogenesis in non-IgE-mediated food allergy and an association with microbiome has not been well established. Previous studies have identified the presence of altered humoral responses to gut bacteria in IgE mediated allergies. Here, we analyzed IgA, IgE and IgG responses to gut bacteria in subjects with either IgE or non-IgE mediated CMA to identify relative proportions of Ig-coated bacteria and characterize unique disease specific microbial signatures. METHODS Multi-parametric flow cytometry analysis was used to identify IgA, IgE and IgG responses to gut bacteria in CMA patients. Cell sorting of Ig coated gut bacteria was subsequently performed followed by high throughput 16S rRNA gene sequencing and specific patterns of humoral responses to gut bacteria assessed in each study group. RESULTS We identified significant alterations in IgA and IgG gut bacterial coating patterns in CMA subjects. Proportions of IgA-coated bacteria were decreased in IgE mediated CMA subjects without atopic dermatitis (ALL) and non-IgE mediated CMA subjects (ENP), compared to healthy controls (CON). In comparison, IgG-coated bacteria was significantly elevated in CMA subjects with atopic dermatitis (AD). Alpha and beta diversities displayed significant differences in IgA-, IgE-, and IgG-coated bacteria in AD and ENP groups. Significant differences in bacteria coated by IgA, IgE and IgG were detected at Phyla, Genus and Species levels and associated bacterial dysbiosis in IgE and non-IgE mediated allergies were identified. Linear discriminant analysis (LDA) effect size (LEFse) revealed unique disease associated bacterial signatures, including several pathogenic bacteria namely Bacteroides fragilis, Ruminococcus gnavus, Eubacterium dolichum, Fusobacterium, Clostridium neonatale and Robinsoniella peoriensis. Receiver operating characteristic curve analysis confirmed the efficiency of using the bacterial signatures identified as biomarkers for disease. CONCLUSIONS Altered IgA and IgG responses to gut bacteria were identified in CMA subjects. The disease-specific responses were associated with alterations in bacterial diversity and concomitant dysbiosis of Ig-coated bacteria in IgE-mediated and non-IgE-mediated CMA pediatric subjects. The identification of pathogenic bacteria uniquely associated with different classes of allergic disease indicates a role of these bacteria in driving disease-specific pathological phenotypes.
Collapse
Affiliation(s)
| | | | - Fariada Badri
- Laboratory of Immunoregulation, Sidra Medicine, Doha, Qatar
| | | | | | | | - Mamoun Elawad
- Department of Gastroenterology, Sidra Medicine, Doha, Qatar
| | - Mehdi Adeli
- Department of Allergy/Immunology, Sidra Medicine, Doha, Qatar
| | - Souhaila Al Khodor
- Microbiome and Host-Microbes Interactions Laboratory, Sidra Medicine, Doha, Qatar
| | | |
Collapse
|
5
|
Cunico D, Giannì G, Scavone S, Buono EV, Caffarelli C. The Relationship Between Asthma and Food Allergies in Children. CHILDREN (BASEL, SWITZERLAND) 2024; 11:1295. [PMID: 39594870 PMCID: PMC11592619 DOI: 10.3390/children11111295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/18/2024] [Accepted: 10/25/2024] [Indexed: 11/28/2024]
Abstract
Asthma and food allergy are two complex allergic diseases with an increasing prevalence in childhood. They share risk factors, including atopic family history, atopic dermatitis, allergen sensitization, and T2 inflammatory pathways. Several studies have shown that in children with a food allergy, the risk of developing asthma, particularly in early childhood, is high. Food allergen intake or the inhalation of aerosolized allergens can induce respiratory symptoms such as bronchospasm. Patients with both conditions have an increased risk of severe asthma exacerbations, hospitalization, and mortality. The current management of clinical food hypersensitivity primarily involves the dietary avoidance of food allergens and the use of self-injectable adrenaline for severe reactions. Poorly controlled asthma limits the prescription of oral immunotherapy to foods, which has emerged as an alternative therapy for managing food allergies. Biological therapies that are effective in severe asthma have been explored for treating food allergies. Omalizumab improves asthma control and, either alone or in combination with oral immunotherapy, increases the threshold of allergen tolerance. Understanding the interplay between asthma and food allergy is crucial for developing successful treatment approaches and ameliorating patient results.
Collapse
Affiliation(s)
| | | | | | | | - Carlo Caffarelli
- Clinica Pediatrica, Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy; (D.C.); (G.G.); (S.S.); (E.V.B.)
| |
Collapse
|
6
|
Castro AM, Sabater C, Gutiérrez-Díaz I, Navarro S, Rodriguez S, Molinos C, Jiménez S, Claver A, Espin B, Domínguez G, Coronel C, Toyos P, Sariego L, Fernández P, Perez D, Margolles A, Díaz JJ, Delgado S. The intestinal microbiome of infants with cow's milk-induced FPIES is enriched in taxa and genes of enterobacteria. J Pediatr Gastroenterol Nutr 2024; 79:841-849. [PMID: 39175183 DOI: 10.1002/jpn3.12356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/31/2024] [Accepted: 08/07/2024] [Indexed: 08/24/2024]
Abstract
OBJECTIVES Food protein-induced enterocolitis syndrome (FPIES) is a severe type of non-IgE (immunoglobulin E)-mediated (NIM) food allergy, with cow's milk (CM) being the most common offending food. The relationship between the gut microbiota and its metabolites with the inflammatory process in infants with CM FPIES is unknown, although evidence suggests a microbial dysbiosis in NIM patients. This study was performed to contribute to the knowledge of the interaction between the gut microbiota and its derived metabolites with the local immune system in feces of infants with CM FPIES at diagnosis. METHODS Twelve infants with CM FPIES and a matched healthy control group were recruited and the gut microbiota was investigated by 16S amplicon and shotgun sequencing. Fatty acids (FAs) were measured by gas chromatography, while immune factors were determined by enzyme-linked immunosorbent assay and Luminex technology. RESULTS A specific pattern of microbiota in the gut of CM FPIES patients was found, characterized by a high abundance of enterobacteria. Also, an intense excretion of FAs in the feces of these infants was observed. Furthermore, correlations were found between fecal bifidobacteria and immune factors. CONCLUSION These fecal determinations may be useful to gain insight into the pathophysiology of this syndrome and should be taken in consideration for future studies of FPIES patients.
Collapse
Affiliation(s)
- Ana M Castro
- MicroHealth Group, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC)/Instituto Biosanitario del Principado de Asturias (ISPA), Villaviciosa, Asturias, Spain
| | - Carlos Sabater
- MicroHealth Group, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC)/Instituto Biosanitario del Principado de Asturias (ISPA), Villaviciosa, Asturias, Spain
| | - Isabel Gutiérrez-Díaz
- MicroHealth Group, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC)/Instituto Biosanitario del Principado de Asturias (ISPA), Villaviciosa, Asturias, Spain
| | - Sandra Navarro
- Primary Care Center Teatinos-Corredoria, Oviedo, Asturias, Spain
| | - Silvia Rodriguez
- Paediatrics Service, Hospital Universitario de San Agustín, Avilés, Asturias, Spain
| | - Cristina Molinos
- Paediatrics Department, Hospital Universitario de Cabueñes, Gijón, Asturias, Spain
| | | | - Angela Claver
- Allergology, Hospital Universitario Dexeus, Barcelona, Spain
| | - Beatriz Espin
- Paediatric Gastroenterology Unit, Hospital Universitario Virgen del Rocío de Sevilla, Sevilla, Spain
| | - Gloria Domínguez
- Gastroenterology and Nutrition Section, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | | | - Paula Toyos
- Paediatric Group, ISPA, Oviedo, Asturias, Spain
| | - Lydia Sariego
- MicroHealth Group, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC)/Instituto Biosanitario del Principado de Asturias (ISPA), Villaviciosa, Asturias, Spain
| | | | - David Perez
- Paediatrics Service, Hospital Universitario de San Agustín, Avilés, Asturias, Spain
| | - Abelardo Margolles
- MicroHealth Group, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC)/Instituto Biosanitario del Principado de Asturias (ISPA), Villaviciosa, Asturias, Spain
| | - Juan J Díaz
- Paediatric Group, ISPA, Oviedo, Asturias, Spain
| | - Susana Delgado
- MicroHealth Group, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC)/Instituto Biosanitario del Principado de Asturias (ISPA), Villaviciosa, Asturias, Spain
| |
Collapse
|
7
|
Nunes de Castilho Santos L. [Differential diagnosis in food allergy]. REVISTA ALERGIA MÉXICO 2023; 70:260-264. [PMID: 38506869 DOI: 10.29262/ram.v70i4.1312] [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: 08/01/2023] [Accepted: 10/29/2023] [Indexed: 03/21/2024] Open
Abstract
It is important to establish the differential diagnosis of food allergy with other disorders, for example: toxic reactions that occur in any person exposed to a sufficient amount of some allergen, and non-toxic reactions that depend on individual susceptibility (food allergy or intolerance). The differential diagnosis is decisive to establish the appropriate treatment. Food intolerance involves adverse reactions to foods without any immunological response involved, and commonly manifests with gastrointestinal symptoms (malaise, abdominal pain or diarrhea). Food allergy is an exaggerated reaction of the immune system, often mediated by IgE, that can trigger serious symptoms (hives, inflammation, respiratory distress, even anaphylaxis). The complex thing is because the symptoms sometimes overlap. To establish an accurate diagnosis, exhaustive clinical evaluation, laboratory tests and, in some cases, controlled provocation tests are required. It is important to understand these distinctions, because treatment and management vary significantly. Food intolerance involves the elimination or reduction of the food that triggers the allergic reaction and requires rigorous measures (complete avoidance of the allergen and availability of epinephrine in cases of severe reactions).
Collapse
Affiliation(s)
- Liziane Nunes de Castilho Santos
- Alergólogo e Inmunólogo; Responsable Técnico y Profesor del sector de Alergia e Inmunología del Instituto Nacional de Salud de la Mujer, del Niño y del Adolescente Fernandes Figueira IFF/Fiocruz,
| |
Collapse
|
8
|
Rojo Gutiérrez MI, Ballesteros González D, Ortiz Durán AK. [Non-IgE-mediated food allergy]. REVISTA ALERGIA MÉXICO 2023; 70:269-279. [PMID: 38506871 DOI: 10.29262/ram.v70i4.1338] [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: 08/01/2023] [Accepted: 10/29/2023] [Indexed: 03/21/2024] Open
Abstract
Food allergy is an immune response to proteins in food. It usually affects 8% of children and 2% of adults in Western countries. Non-IgE-mediated food allergy mainly affects the gastrointestinal tract. Gastrointestinal food allergies are classified, by their underlying pathogenesis, as: IgE-mediated, non-IgE-mediated, or mixed. The symptoms of patients with food protein-induced allergic proctocolitis originate from local inflammation of the distal colon, which causes hematochezia in neonates. It can affect the entire gastrointestinal tract and cause symptoms of intractable emesis, with subsequent metabolic disorders and hypovolemic shock. Food protein-induced enterocolitis syndrome is a non-IgE-mediated allergy that usually appears in childhood, with prolonged repetitive vomiting, starting 1 to 4 hours after ingestion of food. The manifestation in adults is usually triggered by the consumption of shellfish. Atopic diseases affect 40-60% of patients with food protein- induced enterocolitis syndrome, including 40-50% of those with food protein-induced enteropathy and proctocolitis. Probiotics (Lactobacillus GG) can alleviate the symptoms of allergic proctocolitis induced by food proteins, by altering the composition of the intestinal microbiota. Fecal microbiota transplantation (FMT) can change intestinal microecology efficiently compared to food or probiotics.
Collapse
Affiliation(s)
- María Isabel Rojo Gutiérrez
- Alergóloga e Inmunóloga clínica, Máster en Ciencias y Educación; Miembro de la Mesa Directiva de SLAAI; miembro activo del Colegio Mexicano de Inmunología Clínica y Alergia; Directora de Alergología en la Unidad Médica Zúrich, Ciudad de
| | - Diego Ballesteros González
- Médico Cirujano y Partero, Escuela Superior de Medicina, Instituto Politécnico Nacional; Alergia e inmunología clínica, Hospital Juárez de México
| | | |
Collapse
|
9
|
Barni S, Mori F, Giovannini M, Liotti L, Mastrorilli C, Pecoraro L, Saretta F, Castagnoli R, Arasi S, Caminiti L, Gelsomino M, Klain A, del Giudice MM, Novembre E. Allergic Proctocolitis: Literature Review and Proposal of a Diagnostic-Therapeutic Algorithm. Life (Basel) 2023; 13:1824. [PMID: 37763228 PMCID: PMC10533178 DOI: 10.3390/life13091824] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 08/22/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Allergic proctocolitis (AP) is a benign condition, frequent in childhood, that is classified as a non-IgE-mediated food allergy. The prevalence is unknown; however, its frequency appears to be increasing, especially in exclusively breastfed infants. Clinical manifestations typically begin in the first few months of life with the appearance of bright red blood (hematochezia), with or without mucus, in the stool of apparently healthy, thriving infants. Most cases of AP are caused by cow's milk proteins; however, other allergens, such as soy, egg, corn, and wheat, may be potential triggers. Diagnosis is based on the patient's clinical history and on the resolution of signs and symptoms with the elimination of the suspected food antigen from the diet and their reappearance when the food is reintroduced into the diet. The treatment of AP is based on an elimination diet of the trigger food, with resolution of the symptoms within 72-96 h from the beginning of the diet. The prognosis of AP is good; it is a self-limiting condition, because most children can tolerate the trigger food within one year of life, with an excellent long-term prognosis. The purpose of this review is to provide an update on the current knowledge and recommendations in epidemiological, diagnostic, and therapeutic terms to the pediatricians, allergists, and gastroenterologists who may find themselves managing a patient with AP.
Collapse
Affiliation(s)
- Simona Barni
- Allergy Unit, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy; (S.B.); (F.M.); (M.G.); (E.N.)
| | - Francesca Mori
- Allergy Unit, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy; (S.B.); (F.M.); (M.G.); (E.N.)
| | - Mattia Giovannini
- Allergy Unit, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy; (S.B.); (F.M.); (M.G.); (E.N.)
- Department of Health Sciences, University of Florence, 50139 Florence, Italy
| | - Lucia Liotti
- Pediatric Unit, Department of Mother and Child Health, Salesi Children’s Hospital, 60123 Ancona, Italy;
| | - Carla Mastrorilli
- Pediatric and Emergency Department, Pediatric Hospital Giovanni XXIII, AOU Policlinic of Bari, 70126 Bari, Italy;
| | - Luca Pecoraro
- Pediatric Unit, Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, 37126 Verona, Italy;
| | - Francesca Saretta
- Pediatric Department, Latisana-Palmanova Hospital, Azienda Sanitaria Universitaria Friuli Centrale, 33100 Udine, Italy;
| | - Riccardo Castagnoli
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy;
- Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Stefania Arasi
- Translational Research in Pediatric Specialties Area, Division of Allergy, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy;
| | - Lucia Caminiti
- Allergy Unit, Department of Pediatrics, AOU Policlinico Gaetano Martino, 98124 Messina, Italy;
| | - Mariannita Gelsomino
- Department of Life Sciences and Public Health, Pediatric Allergy Unit, University Foundation Policlinico Gemelli IRCCS, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Angela Klain
- Department of Woman, Child and General and Specialized Surgery, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.K.); (M.M.d.G.)
| | - Michele Miraglia del Giudice
- Department of Woman, Child and General and Specialized Surgery, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.K.); (M.M.d.G.)
| | - Elio Novembre
- Allergy Unit, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy; (S.B.); (F.M.); (M.G.); (E.N.)
| |
Collapse
|
10
|
Caparrós E, Cenit MC, Muriel J, Benítez-Páez A, Moreno MV, González-Delgado P, Rubio G, Sanz Y, Fernández J. Intestinal microbiota is modified in pediatric food protein-induced enterocolitis syndrome. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2022; 1:217-224. [PMID: 37779539 PMCID: PMC10510020 DOI: 10.1016/j.jacig.2022.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 07/24/2022] [Accepted: 07/31/2022] [Indexed: 10/03/2023]
Abstract
Background Food protein-induced enterocolitis syndrome (FPIES) is a non-IgE-mediated food hypersensitivity that affects the gastrointestinal system, especially in children, who often present with more severe clinical manifestations than adults do. Although its pathogenesis is poorly understood and biomarkers are still lacking, scientific evidence suggests that gut microbiota may play an important role in the development of FPIES. Objective We aimed to compare the composition of gut microbiota in children with FPIES with that in age- and sex-matched healthy controls. Methods We analyzed the gut microbiota profiles in fecal samples of 17 patients with FPIES (case patients) and 12 age-matched healthy children (controls) by tag sequencing of the 16S ribosomal RNA gene hypervariable V4-V5 regions. Subjects' sociodemographic, clinical, and food diary variables were described and compared between groups by using inferential statistical tests. Nonparametric linear discriminant analysis was performed for intestinal microbiota data. Results Patients with confirmed cases FPIES (n = 17; average patient age, 7.5 ± 3.2 years) and controls without FPIES or any atopy (n = 12, average patient age, 6.9 ± 2.7 years) were included. Fish was the main FPIES-inducing allergen in 65% of the cases. The patients with FPIES showed higher proportions of Lachnospiraceae spp (P < .0286) and a lower proportion of Ruminococcaceae spp (P < .0066), Lactobacillaceae spp (P < .0075), and Leuconostocaceae spp (P < .0173) than the controls. Conclusions Our data clearly show a different gut microbial signature in patients with FPIES, suggesting a new potential avenue for aiding the diagnosis and clinical management of FPIES. Larger studies are needed to confirm these results.
Collapse
Affiliation(s)
- Esther Caparrós
- Clinical Medicine Department, University Miguel Hernández, San Juan de Alicante, Alicante, Spain
| | - María Carmen Cenit
- Microbial Ecology, Nutrition, and Health Research Unit, Institute of Agrochemistry and Food Technology, Paterna, Valencia, Spain
- Foundation for the Promotion of Sanitary and Biomedical Research of Valencian Community, Valencia, Spain
| | - Javier Muriel
- Clinical Medicine Department, University Miguel Hernández, San Juan de Alicante, Alicante, Spain
| | - Alfonso Benítez-Páez
- Microbial Ecology, Nutrition, and Health Research Unit, Institute of Agrochemistry and Food Technology, Paterna, Valencia, Spain
- Host-Microbe Interactions in Metabolic Health Laboratory, Principe Felipe Research Center, Valencia, Spain
| | - María Victoria Moreno
- Clinical Medicine Department, University Miguel Hernández, San Juan de Alicante, Alicante, Spain
| | - Purificación González-Delgado
- Allergy Service, Alicante General University Hospital, Alicante Institute for Health and Biomedical Research, Alicante, Spain
| | - Gonzalo Rubio
- Department of Biochemistry, Molecular Biology “B” and Immunology, University of Murcia, Murcia, Spain
| | - Yolanda Sanz
- Microbial Ecology, Nutrition, and Health Research Unit, Institute of Agrochemistry and Food Technology, Paterna, Valencia, Spain
| | - Javier Fernández
- Clinical Medicine Department, University Miguel Hernández, San Juan de Alicante, Alicante, Spain
- Allergy Service, Alicante General University Hospital, Alicante Institute for Health and Biomedical Research, Alicante, Spain
| |
Collapse
|
11
|
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
|
12
|
Bai J, Zhao X, Zhang M, Xia X, Yang A, Chen H. Gut microbiota: A target for prebiotics and probiotics in the intervention and therapy of food allergy. Crit Rev Food Sci Nutr 2022; 64:3623-3637. [PMID: 36218372 DOI: 10.1080/10408398.2022.2133079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Food allergy has become a major public health problem all over the world. Evidence showed that allergic reactions induced by food proteins often lead to disturbances in the gut microbiota (symbiotic bacteria). Gut microbiota plays an important role in maintaining the balance between intestinal immune tolerance and allergic reactions. Dietary intervention has gradually become an important method for the prevention and treatment of allergic diseases, and changing the composition of gut microbiota through oral intake of prebiotics and probiotics may serve as a new effective adjuvant treatment measure for allergic diseases. In this paper, the main mechanism of food allergy based on intestinal immunity was described firstly. Then, the clinical and experimental evidence showed that different prebiotics and probiotics affect food allergy by changing the structure and composition of gut microbiota was summarized. Moreover, the molecular mechanism in which the gut microbiota and their metabolites may directly or indirectly regulate the immune system or intestinal epithelial barrier function to affect food immune tolerance of host were also reviewed to help in the development of food allergy prevention and treatment strategies based on prebiotics and probiotics.
Collapse
Affiliation(s)
- Jing Bai
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, China
| | - Xiaoli Zhao
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands
| | - Maolin Zhang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, China
| | - Xinlei Xia
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, China
| | - Anshu Yang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, China
| | - Hongbing Chen
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, China
| |
Collapse
|
13
|
陈 顺, 汤 正, 黄 波, 赵 兴, 陈 寿. [Features of intestinal flora in children with food protein-induced proctocolitis based on high-throughput sequencing]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2022; 24:536-542. [PMID: 35644194 PMCID: PMC9154361 DOI: 10.7499/j.issn.1008-8830.2111071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 04/02/2022] [Indexed: 01/25/2023]
Abstract
OBJECTIVES To study the features of intestinal flora in children with food protein-induced proctocolitis (FPIP) by high-throughput sequencing. METHODS A total of 31 children, aged <6 months, who experienced FPIP after exclusive breastfeeding and attended the outpatient service of the Third Affiliated Hospital of Zunyi Medical University from October 2018 to February 2021 were enrolled as the FPIP group. Thirty-one healthy infants were enrolled as the control group. Fecal samples were collected to extract DNA for PCR amplification. High-throughput sequencing was used to perform a bioinformatics analysis of 16S rDNA V3-V4 fragments in fecal samples. RESULTS The diversity analysis of intestinal flora showed that compared with the control group, the FPIP group had a lower Shannon index for diversity (P>0.05) and a significantly higher Chao index for abundance (P<0.01). At the phylum level, the intestinal flora in both groups were composed of Firmicutes, Actinobacteria, Proteobacteria, and Bacteroidetes. Compared with the control group, the FPIP group had a significant reduction in the composition ratio of Actinobacteria (P<0.001) and a significant increase in the composition ratio of Proteobacteria (P<0.05). At the genus level, the intestinal flora in the FPIP group were mainly composed of Escherichia, Clostridium, Enterococcus, Klebsiella, and Bifidobacterium, and the intestinal flora in the control group were mainly composed of Bifidobacterium and Streptococcus. Compared with the control group, the FPIP group had a significant reduction in the composition ratio of Bifidobacterium and Ruminococcus (P<0.05) and significant increases in the composition ratios of Clostridium and Shigella (P<0.05). CONCLUSIONS Compared with the control group, the FPIP group has a reduction in the diversity of intestinal flora and an increase in their abundance, and there are certain differences in several bacterial genera. These results suggest that changes in the composition of intestinal flora at genus level may play an important role in the development and progression of FPIP.
Collapse
Affiliation(s)
- 顺丽 陈
- 遵义医科大学第三附属医院/遵义市第一人民医院儿科,贵州遵义563000
| | - 正珍 汤
- 遵义医科大学第三附属医院/遵义市第一人民医院儿科,贵州遵义563000
| | - 波 黄
- 遵义医科大学第三附属医院/遵义市第一人民医院儿科,贵州遵义563000
| | - 兴艳 赵
- 遵义医科大学第三附属医院/遵义市第一人民医院儿科,贵州遵义563000
| | - 寿珊 陈
- 遵义医科大学第三附属医院/遵义市第一人民医院儿科,贵州遵义563000
| |
Collapse
|
14
|
Du J, Qiao H, Xie D. A prognostic model based on 10 gene signatures associated with intestinal microbiota predicts survival prognosis of esophageal squamous cell adenocarcinoma. ALL LIFE 2022. [DOI: 10.1080/26895293.2022.2046653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Affiliation(s)
- Jiang Du
- Department of Thoracic Surgery, Chinese Medical University Affiliated No. 1 Hospital, Shenyang, People’s Republic of China
| | - Han Qiao
- Clinical Medicine, Chinese Medical University, Shenyang, People’s Republic of China
| | - Dalong Xie
- Department of Anatomy, College of Basic Medicine, China Medical University, Shenyang, People’s Republic of China
| |
Collapse
|
15
|
The gut microbiome-immune axis as a target for nutrition-mediated modulation of food allergy. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2021.05.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
16
|
Fiocchi A, Vickery BP, Wood RA. The use of biologics in food allergy. Clin Exp Allergy 2021; 51:1006-1018. [PMID: 33966304 DOI: 10.1111/cea.13897] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/29/2021] [Accepted: 04/30/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Food allergy continues to pose problems due to its increased frequency and its increasingly high severity. In this context, alongside the traditional avoidance strategies of allergenic foods and desensitization through the cautious progression of exposure to foods in the context of oral immunotherapy (OIT), alternative strategies have made their way in the last decades. We review the possibilities of intervention in food allergy with the use of biological drugs capable of interfering with the synthesis of IgE, with their mechanisms of action, or with complex biological mechanisms that lead to the establishment of a food allergy. METHODS Repeated Entrez PubMed searches using the template algorithm "Food allergy" and "biologics" or "Omalizumab" or "Dupilumab" or "milk desensitization" or "oral tolerance induction" or "oral immunotherapy" or "Etokimab" or "Tezepelumab" or "Quilizumab" or "Ligelizumab" or "Tralokinumab" or "Nemolizumab" or "Mepolizumab" or "Reslizumab" or "Benralizumab". The authors' clinical experience in paediatric allergy units of University hospitals was also drawn upon. RESULTS The landscape in this context has changed dramatically over the past 10 years. We have acquired knowledge mainly on the effect of different types of anti-IgE treatments in poliallergic patients with food allergy, and in patients treated with OIT. However, other mediators are being targeted by specific biologic treatments. Among them, the alarmins Il-33 and TSLP, IL-4 and IL-13, eosinophil-related molecules as IL-6, IL-8, IL-10, IL-12, and mostly IL-5, and integrins involved in the pathogenesis of eosinophilic gastrointestinal diseases (EGIDs), as SIGLEC-8. CONCLUSIONS The ever-better knowledge of the mechanisms of food allergy allowing these developments will improve not only the perspective of patients with the most serious immediate food allergies such as anaphylaxis, but also those of patients with related diseases such as atopic dermatitis, eosinophilic esophagitis, and EGIDs. Biologics are also intended to complement OIT strategies that have developed over the years.
Collapse
Affiliation(s)
| | | | - Robert A Wood
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
17
|
Di Costanzo M, De Paulis N, Biasucci G. Butyrate: A Link between Early Life Nutrition and Gut Microbiome in the Development of Food Allergy. Life (Basel) 2021; 11:life11050384. [PMID: 33922797 PMCID: PMC8146414 DOI: 10.3390/life11050384] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/14/2021] [Accepted: 04/22/2021] [Indexed: 12/11/2022] Open
Abstract
Increased prevalence of food allergies in the last thirty years has been attributed to lifestyle changes in Westernized countries. Among the environmental factors, nutritional factors and their interaction with the gut microbiome in early life are thought to have an important role in the observed epidemiological change. The gut microbiome synthesizes bacterial metabolites, which represent a link among gut microbiome, nutrition, and immune system. The main metabolites produced by gut microbiome are short-chain fatty acids (SCFAs). SCFAs have multiple beneficial effects on human health including protective effects in autoimmune and inflammatory diseases. Among SCFAs, butyrate is essential for maintaining gut immune homeostasis and exerts a pivotal role in immune tolerance with strong anti-inflammatory effects in allergic diseases. Recent findings suggest that butyrate takes part in the development of immunological tolerance to food, especially in the first 1000 days of life. Herein, we provide a critical review of the scientific literature on the role of butyrate for prevention and treatment of food allergies with focus on the complex interplay among early life nutrition, gut microbiome, and immune system.
Collapse
Affiliation(s)
- Margherita Di Costanzo
- Pediatrics and Neonatology Unit, Department of Maternal and Child Health, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy; (N.D.P.); (G.B.)
- Department of Translational Medical Science—Pediatric Section, University “Federico II”, 80131 Naples, Italy
- Correspondence:
| | - Nicoletta De Paulis
- Pediatrics and Neonatology Unit, Department of Maternal and Child Health, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy; (N.D.P.); (G.B.)
| | - Giacomo Biasucci
- Pediatrics and Neonatology Unit, Department of Maternal and Child Health, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy; (N.D.P.); (G.B.)
| |
Collapse
|
18
|
Barni S, Giovannini M, Mori F. Epidemiology of non-IgE-mediated food allergies: what can we learn from that? Curr Opin Allergy Clin Immunol 2021; 21:188-194. [PMID: 33394702 DOI: 10.1097/aci.0000000000000721] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW To underline the main characteristics of the non-Immunoglobulin E (IgE)-mediated food allergies (food protein-induced allergic proctocolitis food protein-induced enteropathy and food protein-induced enterocolitis syndrome ), which are common diseases in primary care and in allergy and gastroenterology specialty practices evaluating children. RECENT FINDINGS Non-IgE-mediated food allergies comprise a spectrum of diseases with peculiar features affecting infants and young children. The most prominent features of these diseases are symptoms that affect mainly the gastrointestinal tract. SUMMARY It is of paramount importance to provide the clinicians with the tools for non-IgE-mediated food allergy recognition in clinical practice to avoid the misdiagnosis with unnecessary laboratory tests and detrimental treatments.
Collapse
Affiliation(s)
- Simona Barni
- Allergy Unit, Department of Pediatrics, Meyer Children's University Hospital, Florence, Italy
| | | | | |
Collapse
|
19
|
Mennini M, Reddel S, Del Chierico F, Gardini S, Quagliariello A, Vernocchi P, Valluzzi RL, Fierro V, Riccardi C, Napolitano T, Fiocchi AG, Putignani L. Gut Microbiota Profile in Children with IgE-Mediated Cow's Milk Allergy and Cow's Milk Sensitization and Probiotic Intestinal Persistence Evaluation. Int J Mol Sci 2021; 22:ijms22041649. [PMID: 33562104 PMCID: PMC7915344 DOI: 10.3390/ijms22041649] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 12/23/2022] Open
Abstract
Food allergy (FA) and, in particular, IgE-mediated cow's milk allergy is associated with compositional and functional changes of gut microbiota. In this study, we compared the gut microbiota of cow's milk allergic (CMA) infants with that of cow's milk sensitized (CMS) infants and Healthy controls. The effect of the intake of a mixture of Bifidobacterium longum subsp. longum BB536, Bifidobacterium breve M-16V and Bifidobacterium longum subsp. infantis M-63 on gut microbiota modulation of CMA infants and probiotic persistence was also investigated. Gut microbiota of CMA infants resulted to be characterized by a dysbiotic status with a prevalence of some bacteria as Haemophilus, Klebsiella, Prevotella, Actinobacillus and Streptococcus. Among the three strains administered, B.longum subsp. infantis colonized the gastrointestinal tract and persisted in the gut microbiota of infants with CMA for 60 days. This colonization was associated with perturbations of the gut microbiota, specifically with the increase of Akkermansia and Ruminococcus. Multi-strain probiotic formulations can be studied for their persistence in the intestine by monitoring specific bacterial probes persistence and exploiting microbiota profiling modulation before the evaluation of their therapeutic effects.
Collapse
Affiliation(s)
- Maurizio Mennini
- Translational Research in Pediatric Specialities Area, Allergy Unit, Bambino Gesù Children’s Hospital, IRCCS, 00147 Rome, Italy; (M.M.); (R.L.V.); (V.F.); (C.R.); (T.N.); (A.G.F.)
| | - Sofia Reddel
- Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00147 Rome, Italy; (S.R.); (F.D.C.); (A.Q.); (P.V.)
| | - Federica Del Chierico
- Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00147 Rome, Italy; (S.R.); (F.D.C.); (A.Q.); (P.V.)
| | | | - Andrea Quagliariello
- Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00147 Rome, Italy; (S.R.); (F.D.C.); (A.Q.); (P.V.)
| | - Pamela Vernocchi
- Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00147 Rome, Italy; (S.R.); (F.D.C.); (A.Q.); (P.V.)
| | - Rocco Luigi Valluzzi
- Translational Research in Pediatric Specialities Area, Allergy Unit, Bambino Gesù Children’s Hospital, IRCCS, 00147 Rome, Italy; (M.M.); (R.L.V.); (V.F.); (C.R.); (T.N.); (A.G.F.)
| | - Vincenzo Fierro
- Translational Research in Pediatric Specialities Area, Allergy Unit, Bambino Gesù Children’s Hospital, IRCCS, 00147 Rome, Italy; (M.M.); (R.L.V.); (V.F.); (C.R.); (T.N.); (A.G.F.)
| | - Carla Riccardi
- Translational Research in Pediatric Specialities Area, Allergy Unit, Bambino Gesù Children’s Hospital, IRCCS, 00147 Rome, Italy; (M.M.); (R.L.V.); (V.F.); (C.R.); (T.N.); (A.G.F.)
| | - Tania Napolitano
- Translational Research in Pediatric Specialities Area, Allergy Unit, Bambino Gesù Children’s Hospital, IRCCS, 00147 Rome, Italy; (M.M.); (R.L.V.); (V.F.); (C.R.); (T.N.); (A.G.F.)
| | - Alessandro Giovanni Fiocchi
- Translational Research in Pediatric Specialities Area, Allergy Unit, Bambino Gesù Children’s Hospital, IRCCS, 00147 Rome, Italy; (M.M.); (R.L.V.); (V.F.); (C.R.); (T.N.); (A.G.F.)
| | - Lorenza Putignani
- Department of Diagnostic and Laboratory Medicine, Unit of Parasitology and Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00147 Rome, Italy
- Correspondence:
| |
Collapse
|
20
|
Bergmann KC, Krause L, Hiller J, Becker S, Kugler S, Tapparo M, Pfaar O, Zuberbier T, Kramer MF, Guethoff S, Graessel A. First evaluation of a symbiotic food supplement in an allergen exposure chamber in birch pollen allergic patients. World Allergy Organ J 2020; 14:100494. [PMID: 33376575 PMCID: PMC7753943 DOI: 10.1016/j.waojou.2020.100494] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/16/2020] [Accepted: 11/16/2020] [Indexed: 12/30/2022] Open
Abstract
Background Allergic rhinitis/rhinoconjunctivitis is the most common immune disease worldwide, but still largely underestimated, underdiagnosed, and undertreated. Dysbiosis and reduced microbial diversity is linked to the development of allergies, and the immunomodulatory effects of pro- and prebiotics might be used to counteract microbiome dysbiosis in allergy. Adequate symbiotic (multi-strain pro-, plus prebiotic) supplementation can be suggested as a complementary approach in the management of allergic rhinitis. Objective The effects of the daily intake of a symbiotic food supplement (combination of Lactobacillus acidophilus NCFM and Bifidobacterium lactis BL-04 with Fructo-Oligosaccharides) for 4 months in birch pollen allergic rhinoconjunctivitis patients were investigated for the first time in an allergen exposure chamber (AEC) allowing standardised, reproducible pollen exposure before and after intake. Methods Eligible patients were exposed to birch pollen (8000 pollen/m³ for 120 min) at the GA2LEN AEC, at baseline (V1) and final visit (V3) outside the season. The Total Symptom Score (TSS) and the scores for nose, eye, bronchial system, and others were evaluated every 10 min during exposure. Other secondary endpoints were the changes in well-being, Peak Nasal Inspiratory Flow (PNIF), lung function parameters, and safety. Co-primary endpoints were differences in Total Nasal Symptom Score (TNSS) and TSS after 120 min of exposure between both visits. Temporal evolution of symptom scores were analysed in an exploratory way using linear mixed effects models. Results 27 patients (mean age 45 years, 15% male) completed the study. Both co-primary endpoints showed significant improvement after intake of the symbiotic. Median TNSS and TSS were decreased 50% and 80% at 120 min (adjusted p-value = 0.025 and p < 0.01 respectively). All four symptom scores and the personal well-being, improved to a clinically relevant extent over time, visible by a weaker increase in symptoms during 120 min of the final birch pollen exposure. No relevant differences were observed for PNIF, PEF, and spirometry. There were no airway obstructions or lung restrictions before and after both exposures. Late phase reactions after exposure were reduced after V3, documenting a better birch pollen tolerability of the patients. The safety and tolerability profile of the symbiotic food supplement was excellent, no adverse events (AEs) were observed. Conclusions This first evaluation of a symbiotic food supplement in an AEC in rhinoconjunctivitis patients with or without asthma induced by birch pollen revealed a significant beneficial effect, harnessing significant improvements of symptoms and well-being while maintaining an excellent safety and tolerability profile.
Collapse
Affiliation(s)
- Karl-Christian Bergmann
- Department for Dermatology and Allergy, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, And Berlin Institute of Health, Berlin, Germany
- ECARF - European Centre for Allergy Research Foundation, Berlin, Germany
| | - Linda Krause
- Institute of Medical Biometry and Epidemiology, University Medical Center Hamburg- Eppendorf, Hamburg, Germany
| | | | - Sylvia Becker
- ECARF - European Centre for Allergy Research Foundation, Berlin, Germany
| | - Sebastian Kugler
- ECARF - European Centre for Allergy Research Foundation, Berlin, Germany
| | | | - Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Rhinology and Allergy, University Hospital Marburg, Philipps-Universität Marburg, Marburg, Germany
| | - Torsten Zuberbier
- Department for Dermatology and Allergy, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, And Berlin Institute of Health, Berlin, Germany
- ECARF - European Centre for Allergy Research Foundation, Berlin, Germany
| | - Matthias F. Kramer
- Bencard Allergie GmbH, Munich, Germany
- Allergy Therapeutics (UK) Ltd, Worthing, UK
| | | | - Anke Graessel
- Bencard Allergie GmbH, Munich, Germany
- Allergy Therapeutics (UK) Ltd, Worthing, UK
- Corresponding author. Leopoldstr. 175, 80804 Muenchen, Germany
| |
Collapse
|
21
|
Di Costanzo M, Carucci L, Berni Canani R, Biasucci G. Gut Microbiome Modulation for Preventing and Treating Pediatric Food Allergies. Int J Mol Sci 2020; 21:ijms21155275. [PMID: 32722378 PMCID: PMC7432728 DOI: 10.3390/ijms21155275] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 12/11/2022] Open
Abstract
The increasing prevalence and severity of pediatric food allergies (FA) demands innovative preventive and therapeutic strategies. Emerging evidence suggests a pivotal role for the gut microbiome in modulating susceptibility to FA. Studies have demonstrated that alteration of gut microbiome could precede FA, and that particular microbial community structures early in life could influence also the disease course. The identification of gut microbiome features in pediatric FA patients is driving new prevention and treatment approaches. This review is focused on the potential role of the gut microbiome as a target for FA prevention and treatment.
Collapse
Affiliation(s)
- Margherita Di Costanzo
- Department of Pediatrics and Neonatology, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy;
- Department of Translational Medical Science-Pediatric Section, University “Federico II”, 80131 Naples, Italy; (L.C.); (R.B.C.)
- ImmunoNutritionLab-CEINGE Advanced Biotechnologies, University “Federico II”, 80131 Naples, Italy
- Correspondence:
| | - Laura Carucci
- Department of Translational Medical Science-Pediatric Section, University “Federico II”, 80131 Naples, Italy; (L.C.); (R.B.C.)
- ImmunoNutritionLab-CEINGE Advanced Biotechnologies, University “Federico II”, 80131 Naples, Italy
| | - Roberto Berni Canani
- Department of Translational Medical Science-Pediatric Section, University “Federico II”, 80131 Naples, Italy; (L.C.); (R.B.C.)
- ImmunoNutritionLab-CEINGE Advanced Biotechnologies, University “Federico II”, 80131 Naples, Italy
- Task Force on Microbiome Studies, University of Naples “Federico II”, 80131 Naples, Italy
- European Laboratory for the Investigation of Food-Induced Diseases, University of Naples “Federico II”, 80131 Naples, Italy
| | - Giacomo Biasucci
- Department of Pediatrics and Neonatology, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy;
| |
Collapse
|
22
|
|