1
|
Davis KL, Claudio-Etienne E, Frischmeyer-Guerrerio PA. Atopic dermatitis and food allergy: More than sensitization. Mucosal Immunol 2024; 17:1128-1140. [PMID: 38906220 PMCID: PMC11471387 DOI: 10.1016/j.mucimm.2024.06.005] [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: 03/27/2024] [Revised: 06/01/2024] [Accepted: 06/13/2024] [Indexed: 06/23/2024]
Abstract
The increased risk of food allergy in infants with atopic dermatitis (AD) has long been recognized; an epidemiologic phenomenon termed "the atopic march." Current literature supports the hypothesis that food antigen exposure through the disrupted skin barrier in AD leads to food antigen-specific immunoglobulin E production and food sensitization. However, there is growing evidence that inflammation in the skin drives intestinal remodeling via circulating inflammatory signals, microbiome alterations, metabolites, and the nervous system. We explore how this skin-gut axis helps to explain the link between AD and food allergy beyond sensitization.
Collapse
Affiliation(s)
- Katelin L Davis
- Food Allergy Research Section, Laboratory of Allergic Diseases, The National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA; Comparative Biomedical Scientist Training Program, The Molecular Pathology Unit, Laboratory of Cancer Biology and Genetics, Center for Cancer Research, The National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Comparative Pathobiology Department, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Estefania Claudio-Etienne
- Food Allergy Research Section, Laboratory of Allergic Diseases, The National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Pamela A Frischmeyer-Guerrerio
- Food Allergy Research Section, Laboratory of Allergic Diseases, The National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
2
|
Mousavian AH, Zare Garizi F, Ghoreshi B, Ketabi S, Eslami S, Ejtahed HS, Qorbani M. The association of infant and mother gut microbiomes with development of allergic diseases in children: a systematic review. J Asthma 2024; 61:1121-1135. [PMID: 38506489 DOI: 10.1080/02770903.2024.2332921] [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: 12/20/2023] [Revised: 03/12/2024] [Accepted: 03/15/2024] [Indexed: 03/21/2024]
Abstract
OBJECTIVE It is believed that gut microbiota alteration leads to both intestinal and non-intestinal diseases in children. Since infants inherit maternal microbiota during pregnancy and lactation, recent studies suggest that changes in maternal microbiota can cause immune disorders as well. This systematic review was designed to assess the association between the child and mother's gut microbiome and allergy development in childhood. DATA SOURCES In this systematic review, international databases including PubMed, Scopus, and ISI/WOS were searched until January 2023 to identify relevant studies. STUDY SELECTIONS Observational studies that analyzed infant or maternal stool microbiome and their association with allergy development in children were included in this study. Data extraction and quality assessment of the included studies were independently conducted by two researchers. RESULTS Of the 1694 papers evaluated, 21 studies examined neonate gut microbiome by analyzing stool samples and six studies examined maternal gut microbiota. A total of 5319 participants were included in this study. Asthma followed by eczema and dermatitis were the most common allergy disorders among children. Urbanization caused a lack of diversity in the bacterial microbiota as well as lower levels of Bifidobacterium and Lachnospira associated with a higher risk of allergy. In contrast, higher levels of Roseburia and Flavonifractor were associated with lower allergy risk. CONCLUSIONS This systematic review shows that gut microbiota may be associated with allergy development. Further studies are required to provide a definitive answer.
Collapse
Affiliation(s)
- Amir-Hossein Mousavian
- Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Fateme Zare Garizi
- Student Research Committee, Alborz University of Medical Sciences, Karaj, Iran
| | - Behnaz Ghoreshi
- Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Siavash Ketabi
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Solat Eslami
- Department of Medical Biotechnology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Hanieh-Sadat Ejtahed
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mostafa Qorbani
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| |
Collapse
|
3
|
Liang Y, Zhang C, Xiong X, Mao X, Sun P, Yue Z, Wang W, Xie R, Zhang G. Alterations of gut microbiome in eosinophilic chronic rhinosinusitis. Eur Arch Otorhinolaryngol 2024:10.1007/s00405-024-08931-3. [PMID: 39212702 DOI: 10.1007/s00405-024-08931-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE A growing body of evidence has elucidated that the gut microbiota has a crucial impact on the pathophysiological process of atopic diseases. Eosinophilic chronic rhinosinusitis with nasal polyps (eCRSwNP) is a local atopic disease of the systemic immune response. Alterations in the gut microbiome in eCRSwNP patients remain largely undefined. METHODS 16S rRNA gene sequencing was performed in a cross-sectional study of 17 eCRSwNP patients, 9 noneCRSwNP patients and 13 healthy controls, and gut microbiota DNA sequencing between each pair of groups was compared using bioinformatic methods. RESULTS Compared with that of healthy controls, the gut microbiomes of eCRSwNP patients were characterised by a distinct overall microbial composition. However, no significant differences were found in the alpha diversity of the gut microbiota between patients and healthy controls. The distinct differences in microbial composition were significantly correlated with the severity of disease. At the genus level, the abundance of Faecalibacterium positively correlated with Lund-Mackay CT scores. Similarly, the abundance of Turicibacter positively correlated with the percentage of tissue eosinophils. CONCLUSIONS We found alterations in the gut microbiome in eCRSwNP patients, and the alterations in the gut microbiome were correlated with the severity of disease.
Collapse
Affiliation(s)
- Yibo Liang
- Department of Otorhinolaryngology Head and Neck Surgery, Institute of Otolaryngology of Tianjin, Key Laboratory of Auditory Speech and Balance Medicine, Key Medical Discipline of Tianjin (Otolaryngology), Tianjin First Central Hospital, Quality Control Centre of Otolaryngology, Tianjin, 300192, China
| | - Chenting Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Institute of Otolaryngology of Tianjin, Key Laboratory of Auditory Speech and Balance Medicine, Key Medical Discipline of Tianjin (Otolaryngology), Tianjin First Central Hospital, Quality Control Centre of Otolaryngology, Tianjin, 300192, China
- The First Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Xin Xiong
- Department of Otorhinolaryngology Head and Neck Surgery, Institute of Otolaryngology of Tianjin, Key Laboratory of Auditory Speech and Balance Medicine, Key Medical Discipline of Tianjin (Otolaryngology), Tianjin First Central Hospital, Quality Control Centre of Otolaryngology, Tianjin, 300192, China
| | - Xiang Mao
- Department of Otorhinolaryngology Head and Neck Surgery, Institute of Otolaryngology of Tianjin, Key Laboratory of Auditory Speech and Balance Medicine, Key Medical Discipline of Tianjin (Otolaryngology), Tianjin First Central Hospital, Quality Control Centre of Otolaryngology, Tianjin, 300192, China
| | - Peiyong Sun
- Department of Otorhinolaryngology Head and Neck Surgery, Institute of Otolaryngology of Tianjin, Key Laboratory of Auditory Speech and Balance Medicine, Key Medical Discipline of Tianjin (Otolaryngology), Tianjin First Central Hospital, Quality Control Centre of Otolaryngology, Tianjin, 300192, China
| | - Zhenzhong Yue
- Department of Otorhinolaryngology Head and Neck Surgery, Institute of Otolaryngology of Tianjin, Key Laboratory of Auditory Speech and Balance Medicine, Key Medical Discipline of Tianjin (Otolaryngology), Tianjin First Central Hospital, Quality Control Centre of Otolaryngology, Tianjin, 300192, China
| | - Wei Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Institute of Otolaryngology of Tianjin, Key Laboratory of Auditory Speech and Balance Medicine, Key Medical Discipline of Tianjin (Otolaryngology), Tianjin First Central Hospital, Quality Control Centre of Otolaryngology, Tianjin, 300192, China
| | - Runxiang Xie
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, 59 Liu Ting Street, Haishu District, Ningbo, 315000, Zhejiang, China.
| | - Guimin Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Institute of Otolaryngology of Tianjin, Key Laboratory of Auditory Speech and Balance Medicine, Key Medical Discipline of Tianjin (Otolaryngology), Tianjin First Central Hospital, Quality Control Centre of Otolaryngology, Tianjin, 300192, China.
| |
Collapse
|
4
|
Moriki D, León ED, García-Gamero G, Jiménez-Hernández N, Artacho A, Pons X, Koumpagioti D, Dinopoulos A, Papaevangelou V, Priftis KN, Douros K, Francino MP. Specific Gut Microbiome Signatures in Children with Cow's Milk Allergy. Nutrients 2024; 16:2752. [PMID: 39203888 PMCID: PMC11357501 DOI: 10.3390/nu16162752] [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: 07/31/2024] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 09/03/2024] Open
Abstract
Although gut dysbiosis is associated with cow's milk allergy (CMA), causality remains uncertain. This study aimed to identify specific bacterial signatures that influence the development and outcome of the disease. We also investigated the effect of hypoallergenic formula (HF) consumption on the gut microbiome of milk-allergic children. 16S rRNA amplicon sequencing was applied to characterize the gut microbiome of 32 milk-allergic children aged 5-12 years and 36 age-matched healthy controls. We showed that the gut microbiome of children with CMA differed significantly from that of healthy children, regardless of whether they consumed cow's milk. Compared to that of healthy cow's milk consumers, it was depleted in Bifidobacterium, Coprococcus catus, Monoglobus, and Lachnospiraceae GCA-900066575, while being enriched in Oscillibacter valericigenes, Negativibacillus massiliensis, and three genera of the Ruminococcaceae family. Of these, only the Ruminococcaceae taxa were also enriched in healthy children not consuming cow's milk. Furthermore, the gut microbiome of children who developed tolerance and had received an HF was similar to that of healthy children, whereas that of children who had not received an HF was significantly different. Our results demonstrate that specific gut microbiome signatures are associated with CMA, which differ from those of dietary milk elimination. Moreover, HF consumption affects the gut microbiome of children who develop tolerance.
Collapse
Affiliation(s)
- Dafni Moriki
- 3rd Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University Hospital, 12462 Athens, Greece
| | - E. Daniel León
- Department of Genomics and Health, Fundación Para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valencia (FISABIO), 46020 Valencia, Spain
| | - Gabriel García-Gamero
- Department of Genomics and Health, Fundación Para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valencia (FISABIO), 46020 Valencia, Spain
| | - Nuria Jiménez-Hernández
- Department of Genomics and Health, Fundación Para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valencia (FISABIO), 46020 Valencia, Spain
| | - Alejandro Artacho
- Department of Genomics and Health, Fundación Para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valencia (FISABIO), 46020 Valencia, Spain
| | - Xavier Pons
- Department of Genomics and Health, Fundación Para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valencia (FISABIO), 46020 Valencia, Spain
| | - Despoina Koumpagioti
- Department of Nursing, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Argirios Dinopoulos
- 3rd Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University Hospital, 12462 Athens, Greece
| | - Vassiliki Papaevangelou
- 3rd Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University Hospital, 12462 Athens, Greece
| | - Kostas N. Priftis
- 3rd Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University Hospital, 12462 Athens, Greece
| | - Konstantinos Douros
- 3rd Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University Hospital, 12462 Athens, Greece
| | - M. 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, 28029 Madrid, Spain
| |
Collapse
|
5
|
Ponda P, Cerise JE, Navetta-Modrov B, Kiehm J, Covelli GM, Weiss J, Lee AT. The age-specific microbiome of children with milk, egg, and peanut allergy. Ann Allergy Asthma Immunol 2024; 133:203-210.e6. [PMID: 38697287 DOI: 10.1016/j.anai.2024.04.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/10/2024] [Accepted: 04/22/2024] [Indexed: 05/04/2024]
Abstract
BACKGROUND Immune regulation by gut microbiota is affected by dysbiosis and may precede food allergy onset. Prior studies lacked comparisons stratified by age and clinical phenotype. OBJECTIVE To assess the microbiome of children with food allergy (<3 years, 3-18 years) compared with similar aged children without food allergy. METHODS A real-world prospective cross-sectional study performed from 2014 to 2019 recruited children highly likely to have milk, egg, or peanut allergy defined by history and serum IgE or confirmed by food challenge. 16S ribosomal RNA sequencing identified stool microbial DNA. Alpha and beta diversity was compared between groups with food allergy and healthy controls stratified by age. Differential abundance for non a priori taxa was accepted at absolute fold-change greater than 2 and q value less than 0.05. RESULTS A total of 70 patients were included (56 with food allergy and 14 healthy controls). Groups were not significantly different in age, gender at birth, race, mode of delivery, breastfeeding duration, or antibiotic exposure. Younger children with food allergy had similar alpha diversity compared with controls. Beta diversity was significantly different by age (P = .001). There was differential abundance of several a priori (P < .05) taxa (including Clostridia) only in younger children. Both a priori (including Coprococcus and Clostridia) and non a priori (q < 0.05) Acidobacteria_Gp15, Aestuariispira, Tindallia, and Desulfitispora were significant in older children with food allergy, especially with peanut allergy. CONCLUSION Dysbiosis associates with food allergy, most prominent in older children with peanut allergy. Younger children with and without food allergy have fewer differences in gut microbiota. This correlates with clinical observations of persistence of peanut allergy and improved efficacy and safety of oral immunotherapy in younger children. Age younger than 3 years should be considered when initiating therapeutic interventions.
Collapse
Affiliation(s)
- Punita Ponda
- Northwell, New Hyde Park, New York; Division of Allergy and Immunology, Cohen Children's Medical Center, New Hyde Park, New York.
| | - Jane E Cerise
- Biostatistics Unit, Office of Academic Affairs, Northwell Health, New Hyde Park, New York
| | - Brianne Navetta-Modrov
- Department of Medicine, Stony Brook University School of Medicine, Stony Brook, New York
| | - Jamie Kiehm
- Los Angeles County Department of Health Services, Los Angeles, California
| | - Grace M Covelli
- Walter Reed National Military Medical Center, Bethesda, Maryland
| | - Jared Weiss
- Department of Psychiatry, New York University School of Medicine, New York, New York
| | - Annette T Lee
- Northwell, New Hyde Park, New York; Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York
| |
Collapse
|
6
|
Delgado Dolset MI, Pablo-Torres C, Contreras N, Couto-Rodríguez A, Escolar-Peña A, Graña-Castro O, Izquierdo E, López-Rodríguez JC, Macías-Camero A, Pérez-Gordo M, Villaseñor A, Zubeldia-Varela E, Barber D, Escribese MM. Severe Allergy as a Chronic Inflammatory Condition From a Systems Biology Perspective. Clin Exp Allergy 2024; 54:550-584. [PMID: 38938054 DOI: 10.1111/cea.14517] [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: 01/17/2024] [Revised: 05/14/2024] [Accepted: 05/26/2024] [Indexed: 06/29/2024]
Abstract
Persistent and unresolved inflammation is a common underlying factor observed in several and seemingly unrelated human diseases, including cardiovascular and neurodegenerative diseases. Particularly, in atopic conditions, acute inflammatory responses such as those triggered by insect venom, food or drug allergies possess also a life-threatening potential. However, respiratory allergies predominantly exhibit late immune responses associated with chronic inflammation, that can eventually progress into a severe phenotype displaying similar features as those observed in other chronic inflammatory diseases, as is the case of uncontrolled severe asthma. This review aims to explore the different facets and systems involved in chronic allergic inflammation, including processes such as tissue remodelling and immune cell dysregulation, as well as genetic, metabolic and microbiota alterations, which are common to other inflammatory conditions. Our goal here was to deepen on the understanding of an entangled disease as is chronic allergic inflammation and expose potential avenues for the development of better diagnostic and intervention strategies.
Collapse
Affiliation(s)
- M I Delgado Dolset
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - C Pablo-Torres
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - N Contreras
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - A Couto-Rodríguez
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - A Escolar-Peña
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - O Graña-Castro
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - E Izquierdo
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - J C López-Rodríguez
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - A Macías-Camero
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - M Pérez-Gordo
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - A Villaseñor
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - E Zubeldia-Varela
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - D Barber
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - M M Escribese
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| |
Collapse
|
7
|
Jiang S, Cai M, Li D, Chen X, Chen X, Huang Q, Zhong C, Zheng X, Zhou D, Chen Z, Zhang L, Ching JY, Chen A, Lu S, Zhang L, Hu L, Liao Y, Li Y, He Z, Wu J, Huo H, Liang Y, Li W, Zou Y, Luo W, Ng SC, Chan FK, Chen X, Deng Y. Association of breast milk-derived arachidonic acid-induced infant gut dysbiosis with the onset of atopic dermatitis. Gut 2024:gutjnl-2024-332407. [PMID: 39084687 DOI: 10.1136/gutjnl-2024-332407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 06/27/2024] [Indexed: 08/02/2024]
Abstract
OBJECTIVE The specific breast milk-derived metabolites that mediate host-microbiota interactions and contribute to the onset of atopic dermatitis (AD) remain unknown and require further investigation. DESIGN We enrolled 250 mother-infant pairs and collected 978 longitudinal faecal samples from infants from birth to 6 months of age, along with 243 maternal faecal samples for metagenomics. Concurrently, 239 corresponding breast milk samples were analysed for metabolomics. Animal and cellular experiments were conducted to validate the bioinformatics findings. RESULTS The clinical findings suggested that a decrease in daily breastfeeding duration was associated with a reduced incidence of AD. This observation inspired us to investigate the effects of breast milk-derived fatty acids. We found that high concentrations of arachidonic acid (AA), but not eicosapentaenoic acid (EPA) or docosahexaenoic acid, induced gut dysbiosis in infants. Further investigation revealed that four specific bacteria degraded mannan into mannose, consequently enhancing the mannan-dependent biosynthesis of O-antigen and lipopolysaccharide. Correlation analysis confirmed that in infants with AD, the abundance of Escherichia coli under high AA concentrations was positively correlated with some microbial pathways (eg, 'GDP-mannose-derived O-antigen and lipopolysaccharide biosynthesis'). These findings are consistent with those of the animal studies. Additionally, AA, but not EPA, disrupted the ratio of CD4/CD8 cells, increased skin lesion area and enhanced the proportion of peripheral Th2 cells. It also promoted IgE secretion and the biosynthesis of prostaglandins and leukotrienes in BALB/c mice fed AA following ovalbumin immunostimulation. Moreover, AA significantly increased IL-4 secretion in HaCaT cells costimulated with TNF-α and INF-γ. CONCLUSIONS This study demonstrates that AA is intimately linked to the onset of AD via gut dysbiosis.
Collapse
Affiliation(s)
- Suhua Jiang
- Department of paediatrics, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Mengyun Cai
- Institute of translational medicine, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Dingru Li
- Institute of translational medicine, The First People's Hospital of Foshan, Foshan, People's Republic of China
- South China University of Technology School of Biology and Biological Engineering, Guangzhou, Guangdong, People's Republic of China
| | - Xiangping Chen
- Institute of translational medicine, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Xiaoqian Chen
- Department of paediatrics, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Qitao Huang
- Department of obstetrics, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Caimei Zhong
- Department of Dermatology, Shunde District Center for Prevention and Cure of Chronic Diseases, Foshan, China
| | - Xiufeng Zheng
- Department of Dermatology, Shunde Hospital, Southern Medical University, Lunjiao, Shunde, Foshan, People's Republic of China
| | - Dan Zhou
- Department of Breast Surgery, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Zhiyan Chen
- Department of Breast Surgery, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Lin Zhang
- Microbiota I-Center (MagIC), The Chinese University of Hong Kong, Hong Kong SAR, People's Republic of China
- Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR, People's Republic of China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, People's Republic of China
- Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinse University of Hong Kong, Hong Kong SAR, People's Republic of China
| | - Jessica Yl Ching
- Microbiota I-Center (MagIC), The Chinese University of Hong Kong, Hong Kong SAR, People's Republic of China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, People's Republic of China
| | - Ailing Chen
- Department of paediatrics, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Shaoxia Lu
- Department of obstetrics, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Lifang Zhang
- Institute of translational medicine, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Ling Hu
- Institute of translational medicine, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Yan Liao
- Department of obstetrics, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Ying Li
- Department of paediatrics, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Zhihua He
- Department of obstetrics, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Jingjing Wu
- Department of obstetrics, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Huiyi Huo
- Department of paediatrics, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Yongqi Liang
- Department of paediatrics, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Wanwen Li
- Department of paediatrics, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Yanli Zou
- The Second People's Hospital of Foshan, Foshan, People's Republic of China
| | - Wei Luo
- Institute of translational medicine, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Siew C Ng
- Microbiota I-Center (MagIC), The Chinese University of Hong Kong, Hong Kong SAR, People's Republic of China
- Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR, People's Republic of China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, People's Republic of China
- Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinse University of Hong Kong, Hong Kong SAR, People's Republic of China
| | - Francis Kl Chan
- Microbiota I-Center (MagIC), The Chinese University of Hong Kong, Hong Kong SAR, People's Republic of China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, People's Republic of China
| | - Xia Chen
- Central Laboratory of the Medical Research Center, The First Affiliated Hospital of Ningbo University, Ningbo, People's Republic of China
| | - Yuhua Deng
- Institute of translational medicine, The First People's Hospital of Foshan, Foshan, People's Republic of China
| |
Collapse
|
8
|
Pirker AL, Vogl T. Development of systemic and mucosal immune responses against gut microbiota in early life and implications for the onset of allergies. FRONTIERS IN ALLERGY 2024; 5:1439303. [PMID: 39086886 PMCID: PMC11288972 DOI: 10.3389/falgy.2024.1439303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 07/05/2024] [Indexed: 08/02/2024] Open
Abstract
The early microbial colonization of human mucosal surfaces is essential for the development of the host immune system. Already during pregnancy, the unborn child is prepared for the postnatal influx of commensals and pathogens via maternal antibodies, and after birth this protection is continued with antibodies in breast milk. During this critical window of time, which extends from pregnancy to the first year of life, each encounter with a microorganism can influence children's immune response and can have a lifelong impact on their life. For example, there are numerous links between the development of allergies and an altered gut microbiome. However, the exact mechanisms behind microbial influences, also extending to how viruses influence host-microbe interactions, are incompletely understood. In this review, we address the impact of infants' first microbial encounters, how the immune system develops to interact with gut microbiota, and summarize how an altered immune response could be implied in allergies.
Collapse
Affiliation(s)
| | - Thomas Vogl
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
9
|
Lin N, Chi H, Guo Q, Liu Z, Ni L. Notch Signaling Inhibition Alleviates Allergies Caused by Antarctic Krill Tropomyosin through Improving Th1/Th2 Imbalance and Modulating Gut Microbiota. Foods 2024; 13:1144. [PMID: 38672818 PMCID: PMC11048830 DOI: 10.3390/foods13081144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/19/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
Antarctic krill tropomyosin (AkTM) has been shown in mice to cause IgE-mediated food allergy. The objective of this work was to investigate the role of Notch signaling in AkTM-sensitized mice, as well as to determine the changes in gut microbiota composition and short-chain fatty acids (SCFAs) in the allergic mice. An AkTM-induced food allergy mouse model was built and N-[N-(3,5-difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT) was used as an γ-secretase inhibitor to inhibit the activation of Notch signaling. Food allergy indices, some key transcription factors, histologic alterations in the small intestine, and changes in gut microbiota composition were examined. The results showed that DAPT inhibited Notch signaling, which reduced AkTM-specific IgE, suppressed mast cell degranulation, decreased IL-4 but increased IFN-γ production, and alleviated allergic symptoms. Quantitative real-time PCR and Western blotting analyses revealed that expressions of Hes-1, Gata3, and IL-4 were down-regulated after DAPT treatment, accompanied by increases in T-bet and IFN-γ, indicating that Notch signaling was active in AkTM-sensitized mice and blocking it could reverse the Th1/Th2 imbalance. Expressions of key transcription factors revealed that Notch signaling could promote Th2 cell differentiation in sensitized mice. Furthermore, 16S rRNA sequencing results revealed that AkTM could alter the diversity and composition of gut microbiota in mice, leading to increases in inflammation-inducing bacteria such as Enterococcus and Escherichia-Shigella. Correlation analysis indicated that reduced SCFA concentrations in AkTM-allergic mice may be related to decreases in certain SCFA-producing bacteria, such as Clostridia_UCG-014. The changes in gut microbiota and SCFAs could be partially restored by DAPT treatment. Our findings showed that inhibiting Notch signaling could alleviate AkTM-induced food allergy by correcting Th1/Th2 imbalance and modulating the gut microbiota.
Collapse
Affiliation(s)
- Na Lin
- East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai 200090, China; (N.L.); (Q.G.); (L.N.)
| | - Hai Chi
- East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai 200090, China; (N.L.); (Q.G.); (L.N.)
- College of Food Science and Engineering, Dalian Ocean University, Dalian 116023, China
| | - Quanyou Guo
- East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai 200090, China; (N.L.); (Q.G.); (L.N.)
| | - Zhidong Liu
- East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai 200090, China; (N.L.); (Q.G.); (L.N.)
| | - Ling Ni
- East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai 200090, China; (N.L.); (Q.G.); (L.N.)
| |
Collapse
|
10
|
Cho H, Kim J, Kim S, Jeong HI, Kwon M, Kim HM, Shim JS, Kim K, Baek J, Kyung Y, Choi SJ, Oh SY, Bae J, Won HH, Kim J, Ahn K. Postpartum Maternal Anxiety Affects the Development of Food Allergy Through Dietary and Gut Microbial Diversity During Early Infancy. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2024; 16:154-167. [PMID: 38528383 DOI: 10.4168/aair.2024.16.2.154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 11/29/2023] [Accepted: 12/25/2023] [Indexed: 03/27/2024]
Abstract
PURPOSE We aimed to investigate the mediating factors between maternal anxiety and the development of food allergy (FA) in children until 2 years from birth. METHODS In this longitudinal cohort of 122 mother-child dyads from pregnancy to 24 months of age, we regularly surveyed maternal psychological states, infant feeding data, and allergic symptoms and collected stool samples at 6 months of age for microbiome analysis. Considering the temporal order of data collection, we investigated serial mediating effects and indirect effects among maternal anxiety, dietary diversity (DD), gut microbial diversity, and FA using structural equation modeling. RESULTS Among the 122 infants, 15 (12.3%) were diagnosed with FA. Increased maternal anxiety between 3 and 6 months after delivery was associated with a lower DD score. Infants with low DD at 4 months showed low gut microbial richness, which was associated with FA development. When the infants were grouped into 4 subtypes, using consensus clustering of 13 gut bacteria significantly associated with maternal anxiety and DD, Prevotella, Eubacterium, Clostridiales and Lachnospiraceae were more abundant in the group with lower FA occurrence. CONCLUSIONS Postpartum maternal anxiety, mediated by reduced DD and gut microbial diversity, may be a risk factor for the development of FA in infants during the first 2 years of life.
Collapse
Affiliation(s)
- Hyunbin Cho
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea
| | - Jiwon Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sukyung Kim
- Department of Pediatrics, Hallym University Dongtan Sacred Heart Hospital, Hallym University School of Medicine, Hwaseong, Korea
| | - Hye-In Jeong
- Department of Pediatrics, Eulji University Hospital, Eulji University School of Medicine, Seoul, Korea
| | - Mijeong Kwon
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hyun Mi Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ji Sun Shim
- Statistics and Data Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Kyunga Kim
- Statistics and Data Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Jihyun Baek
- Department of Psychology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yechan Kyung
- Department of Pediatrics, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Korea
| | - Suk-Joo Choi
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Soo-Young Oh
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jaewoong Bae
- R&D Institute, BioEleven Co., Ltd., Seoul, Korea
| | - Hong-Hee Won
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea.
| | - Jihyun Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Seoul, Korea.
| | - Kangmo Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Seoul, Korea
| |
Collapse
|
11
|
Zubeldia-Varela E, Ibáñez-Sandín MD, Gomez-Casado C, Pérez-Gordo M. Allergy-associated biomarkers in early life identified by Omics techniques. FRONTIERS IN ALLERGY 2024; 5:1359142. [PMID: 38464396 PMCID: PMC10920277 DOI: 10.3389/falgy.2024.1359142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/12/2024] [Indexed: 03/12/2024] Open
Abstract
The prevalence and severity of allergic diseases have increased over the last 30 years. Understanding the mechanisms responsible for these diseases is a major challenge in current allergology, as it is crucial for the transition towards precision medicine, which encompasses predictive, preventive, and personalized strategies. The urge to identify predictive biomarkers of allergy at early stages of life is crucial, especially in the context of major allergic diseases such as food allergy and atopic dermatitis. Identifying these biomarkers could enhance our understanding of the immature immune responses, improve allergy handling at early ages and pave the way for preventive and therapeutic approaches. This minireview aims to explore the relevance of three biomarker categories (proteome, microbiome, and metabolome) in early life. First, levels of some proteins emerge as potential indicators of mucosal health and metabolic status in certain allergic diseases. Second, bacterial taxonomy provides insight into the composition of the microbiota through high-throughput sequencing methods. Finally, metabolites, representing the end products of bacterial and host metabolic activity, serve as early indicators of changes in microbiota and host metabolism. This information could help to develop an extensive identification of biomarkers in AD and FA and their potential in translational personalized medicine in early life.
Collapse
Affiliation(s)
- Elisa Zubeldia-Varela
- Institute of Applied Molecular Medicine Nemesio Díez (IMMA), Department of Basic Medical Sciences, Facultad de Medicina. Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - María Dolores Ibáñez-Sandín
- Department of Allergy, H. Infantil Universitario Niño Jesús, FibHNJ, ARADyAL- RETICs Instituto de Salud Carlos III, IIS-P, Madrid, Spain
| | - Cristina Gomez-Casado
- Department of Dermatology, University Hospital Duesseldorf, Heinrich-Heine University, Duesseldorf, Germany
| | - Marina Pérez-Gordo
- Institute of Applied Molecular Medicine Nemesio Díez (IMMA), Department of Basic Medical Sciences, Facultad de Medicina. Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| |
Collapse
|
12
|
Suprunowicz M, Tomaszek N, Urbaniak A, Zackiewicz K, Modzelewski S, Waszkiewicz N. Between Dysbiosis, Maternal Immune Activation and Autism: Is There a Common Pathway? Nutrients 2024; 16:549. [PMID: 38398873 PMCID: PMC10891846 DOI: 10.3390/nu16040549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/05/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
Autism spectrum disorder (ASD) is a neuropsychiatric condition characterized by impaired social interactions and repetitive stereotyped behaviors. Growing evidence highlights an important role of the gut-brain-microbiome axis in the pathogenesis of ASD. Research indicates an abnormal composition of the gut microbiome and the potential involvement of bacterial molecules in neuroinflammation and brain development disruptions. Concurrently, attention is directed towards the role of short-chain fatty acids (SCFAs) and impaired intestinal tightness. This comprehensive review emphasizes the potential impact of maternal gut microbiota changes on the development of autism in children, especially considering maternal immune activation (MIA). The following paper evaluates the impact of the birth route on the colonization of the child with bacteria in the first weeks of life. Furthermore, it explores the role of pro-inflammatory cytokines, such as IL-6 and IL-17a and mother's obesity as potentially environmental factors of ASD. The purpose of this review is to advance our understanding of ASD pathogenesis, while also searching for the positive implications of the latest therapies, such as probiotics, prebiotics or fecal microbiota transplantation, targeting the gut microbiota and reducing inflammation. This review aims to provide valuable insights that could instruct future studies and treatments for individuals affected by ASD.
Collapse
Affiliation(s)
| | | | | | | | - Stefan Modzelewski
- Department of Psychiatry, Medical University of Bialystok, pl. Wołodyjowskiego 2, 15-272 Białystok, Poland; (M.S.); (N.T.); (A.U.); (K.Z.); (N.W.)
| | | |
Collapse
|
13
|
Ma Y, Deng Y, Shao T, Cui Y, Shen Y. Causal effects of gut microbiota in the development of lung cancer and its histological subtypes: A Mendelian randomization study. Thorac Cancer 2024; 15:486-495. [PMID: 38214421 PMCID: PMC10883858 DOI: 10.1111/1759-7714.15220] [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: 10/25/2023] [Revised: 12/29/2023] [Accepted: 01/02/2024] [Indexed: 01/13/2024] Open
Abstract
BACKGROUND Numerous studies have characterized the gut microbiome (GM) in lung cancer (LC). Yet, the causality between GM and LC and its subtypes remain uncharacterized. METHODS Two-sample Mendelian randomization (MR) was designed to investigate the causal relationship between the GM and LC and its subtypes, using publicly available summary data of genome-wide association studies. The researchers ran two groups of MR analyses, including the genome-wide statistical significance threshold (5 × 10-8 ) and the locus-wide significance level (1 × 10-5 ). RESULTS Using MR analysis, we ascertained 42 groups of GM that are intimately linked to LC and its subtypes at the locus-wide significance level. Of the 42 groups, 12 were in LC, nine in non-small cell lung cancer (NSCLC), six in small cell lung cancer (SCLC), two in lung adenocarcinomas, and 13 in lung squamous carcinomas. After false discovery rate correction, we still found a remarkable causal interaction between the Eubacterium ruminantium group and SCLC. Moreover, five groups of GM closely linked to LC and its subtypes were recognised at the genome-wide statistical significance threshold. This finding included one group each in LC, NSCLC and SCLC, two groups in lung adenocarcinoma and none in lung squamous carcinoma. None of the foregoing findings were heterogeneous or horizontal pleiotropy. Reverse MR revealed that genetic susceptibility to LC and its subtypes caused significant changes in three groups of GM. CONCLUSION Our findings substantiate the causality between GM and LC and its subtypes. This study offers fresh insights into the function of GM in mediating the progression of LC.
Collapse
Affiliation(s)
- Yunlei Ma
- Department of Thoracic Surgery, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
| | - Yuqing Deng
- Department of Thoracic Surgery, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
| | - Tingting Shao
- Department of Thoracic Surgery, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Yong Cui
- Department of Thoracic Surgery, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
| | - Yefeng Shen
- Department of Thoracic Surgery, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
14
|
Fredriksson E, Bodén S, Domellöf M, West CE. Fruit Pouch Consumption Does Not Associate with Early Manifestations of Allergic Disease. Nutrients 2023; 15:4318. [PMID: 37892394 PMCID: PMC10609626 DOI: 10.3390/nu15204318] [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: 09/18/2023] [Revised: 10/05/2023] [Accepted: 10/07/2023] [Indexed: 10/29/2023] Open
Abstract
Consumption of acidic fruit pouches in infancy may damage the epithelial barrier in the gastrointestinal tract and is suggested to increase allergy risk. We aimed to explore if a high fruit pouch consumption is associated with a higher incidence of early allergic manifestations. We included 2959 parent-child dyads from the Swedish prospective, population-based NorthPop birth cohort study with parentally reported data on frequency of fruit pouch consumption at 9 months of age, as well as parentally reported eczema, wheeze, physician-diagnosed asthma, and food allergy in the first 18 months of life. Immunoglobulin E levels (IgE) in serum (n = 1792), as response to a food mix and an inhalant mix, were determined at age 18 months. Compared with no consumption, daily consumption of one or more pouches at 9 months of age was associated with inhalant sensitization (odds ratio (OR) 2.27, 95% confidence interval (CI) 1.06-4.87, n = 1792) but did not remain significant in the multivariable adjusted model (aOR 2.08, 95% CI 0.95-4.53, n = 1679). There were no associations between fruit pouch consumption and allergic manifestations at this young age. This study suggests that fruit pouch consumption is not associated with allergic phenotypes or IgE sensitization in early childhood.
Collapse
Affiliation(s)
| | | | | | - Christina E. West
- Department of Clinical Sciences, Pediatrics, Umeå University, SE 90185 Umeå, Sweden; (E.F.); (S.B.); (M.D.)
| |
Collapse
|
15
|
Chen C, Sang Z, Xie Q, Xue W. Effects of hazelnut protein isolate-induced food allergy on the gut microenvironment in a BALB/c mouse model. Food Funct 2023; 14:8761-8774. [PMID: 37718731 DOI: 10.1039/d3fo02324a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
Hazelnuts are reported as among the nuts that cause severe allergic reactions. However, few systematic studies exist on the changes in the gut microenvironment following hazelnut allergy. This study focused on the effects of hazelnut allergy on the duodenum, jejunum, ileum and colon microenvironment in vivo. We established a hazelnut protein isolate (HPI)-allergic mouse model, which was distinguished by the visible allergy symptoms, dropped temperatures and enhanced allergic inflammatory factor levels in serum, such as HPI-specific immunoglobulin E (sIgE), sIgG2a, interleukin-4, histamine, mouse mast cell protease-1, TNF-α, monocyte chemotactic protein-1 and lipopolysaccharide. For HPI sensitized mice, aggravated mast cell degranulation, severe morphologic damage and inflammatory cell infiltration were observed in the duodenum, jejunum, ileum, and colon, while goblet cell numbers were reduced in the duodenum, jejunum and ileum. Secretory IgA of the jejunum and tight junctions of the duodenum and jejunum were decreased significantly after HPI sensitization. There was no remarkable difference in the pH values of small intestinal contents, but the pH values of colonic contents were elevated, which was due to the decreased short-chain fatty acids (mainly acetate, propionate and butyrate) in the colon. The antioxidant capacity of both large and small intestinal contents declined after HPI sensitization, as evidenced by the increased malondialdehyde and decreased superoxide dismutase activity. HPI sensitization induced gut microbiota dysbiosis with decreased α diversity and altered β diversity in colonic contents. Spearman correlation analysis indicated that the increased characteristic genera, namely Bacteroides, Lactobacillus, Alloprevotella, Erysipelatoclostridium, Parabacteroides, and Helicobacter, played potentially synergistic roles in promoting allergy and gut microenvironment dysregulation.
Collapse
Affiliation(s)
- Chen Chen
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Ziqing Sang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Qiang Xie
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Wentong Xue
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| |
Collapse
|
16
|
Patumcharoenpol P, Kingkaw A, Nakphaichit M, Chatchatee P, Suratannon N, Panagiotou G, Vongsangnak W. Exploring Longitudinal Gut Microbiome towards Metabolic Functional Changes Associated in Atopic Dermatitis in Early Childhood. BIOLOGY 2023; 12:1262. [PMID: 37759661 PMCID: PMC10525566 DOI: 10.3390/biology12091262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023]
Abstract
Atopic dermatitis (AD) is a prevalent inflammatory skin disease that has been associated with changes in gut microbial composition in early life. However, there are limited longitudinal studies examining the gut microbiome in AD. This study aimed to explore taxonomy and metabolic functions across longitudinal gut microbiomes associated with AD in early childhood from 9 to 30 months of age using integrative data analysis within the Thai population. Our analysis revealed that gut microbiome diversity was not different between healthy and AD groups; however, significant taxonomic differences were observed. Key gut bacteria with short-chain fatty acids (SCFAs) production potentials, such as Anaerostipes, Butyricicoccus, Ruminococcus, and Lactobacillus species, showed a higher abundance in the AD group. In addition, metabolic alterations between the healthy and AD groups associated with vitamin production and host immune response, such as biosynthesis of menaquinol, succinate, and (Kdo)2-lipid A, were observed. This study serves as the first framework for monitoring longitudinal microbial imbalances and metabolic functions associated with allergic diseases in Thai children during early childhood.
Collapse
Affiliation(s)
- Preecha Patumcharoenpol
- Interdisciplinary Graduate Program in Bioscience, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand; (P.P.); (A.K.)
| | - Amornthep Kingkaw
- Interdisciplinary Graduate Program in Bioscience, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand; (P.P.); (A.K.)
| | - Massalin Nakphaichit
- Department of Biotechnology, Faculty of Agro-Industry, Kasetsart University, Bangkok 10900, Thailand;
| | - Pantipa Chatchatee
- Center of Excellence for Allergy and Clinical Immunology, Division of Allergy, Immunology and Rheumatology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand;
- King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok 10330, Thailand
| | - Narissara Suratannon
- Center of Excellence for Allergy and Clinical Immunology, Division of Allergy, Immunology and Rheumatology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand;
- King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok 10330, Thailand
| | - Gianni Panagiotou
- Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology–Hans Knöll Institute, 07745 Jena, Germany;
- Faculty of Biological Sciences, Friedrich Schiller University, 07743 Jena, Germany
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Wanwipa Vongsangnak
- Department of Zoology, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
- Omics Center for Agriculture, Bioresources, Food, and Health, Kasetsart University (OmiKU), Bangkok 10900, Thailand
| |
Collapse
|
17
|
Deng Y, Wang J, Xie G, Zou G, Li S, Zhang J, Cai W, Xu J. Correlation between gut microbiota and the development of Graves' disease: A prospective study. iScience 2023; 26:107188. [PMID: 37485373 PMCID: PMC10362358 DOI: 10.1016/j.isci.2023.107188] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/19/2023] [Accepted: 06/16/2023] [Indexed: 07/25/2023] Open
Abstract
The association between gut microbiota and development of Graves' disease (GD) remains unclear. This study aimed to profile the gut microbiota of 65 patients newly diagnosed with GD before and after treatment and 33 physical examination personnel via 16S rRNA sequencing. Significant differences in the gut microbiota composition were observed between the two groups, showing relative bacterial abundances of 1 class, 1 order, 5 families, and 14 genera. After treatment, the abundance of the significantly enriched biota in the GD group decreased considerably, whereas that of the previously decreased biota increased considerably. Further, interleukin-17 levels decreased significantly. The random forest method was used to identify 12 genera that can distinguish patients with GD from healthy controls. Our study revealed that the gut microbiota of patients with GD exhibit unique characteristics compared with that of healthy individuals, which may be related to an imbalance in the immune system and gut microbiota.
Collapse
Affiliation(s)
- Yuanyuan Deng
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang 330006, People's Republic of China
- Jiangxi Clinical Research Center for Endocrine and Metabolic Disease, Nanchang 330006, People's Republic of China
- Jiangxi Branch of National Clinical Research Center for Metabolic Disease, Nanchang 330006, People’s Republic of China
| | - Jiao Wang
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang 330006, People's Republic of China
- Jiangxi Clinical Research Center for Endocrine and Metabolic Disease, Nanchang 330006, People's Republic of China
- Jiangxi Branch of National Clinical Research Center for Metabolic Disease, Nanchang 330006, People’s Republic of China
| | - Guijiao Xie
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang 330006, People's Republic of China
| | - Guilin Zou
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang 330006, People's Republic of China
- Jiangxi Clinical Research Center for Endocrine and Metabolic Disease, Nanchang 330006, People's Republic of China
- Jiangxi Branch of National Clinical Research Center for Metabolic Disease, Nanchang 330006, People’s Republic of China
| | - Shanshan Li
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang 330006, People's Republic of China
| | - Jie Zhang
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang 330006, People's Republic of China
| | - Wei Cai
- Department of Medical Genetics and Cell Biology, Medical College of Nanchang University, Nanchang 330006, People’s Republic of China
| | - Jixiong Xu
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang 330006, People's Republic of China
- Jiangxi Clinical Research Center for Endocrine and Metabolic Disease, Nanchang 330006, People's Republic of China
- Jiangxi Branch of National Clinical Research Center for Metabolic Disease, Nanchang 330006, People’s Republic of China
| |
Collapse
|
18
|
Liu K, Cai Y, Song K, Yuan R, Zou J. Clarifying the effect of gut microbiota on allergic conjunctivitis risk is instrumental for predictive, preventive, and personalized medicine: a Mendelian randomization analysis. EPMA J 2023; 14:235-248. [PMID: 37275551 PMCID: PMC10201039 DOI: 10.1007/s13167-023-00321-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 05/03/2023] [Indexed: 06/07/2023]
Abstract
Background Allergic conjunctivitis is an ocular immune disease which affects the conjunctiva, eyelids, and cornea. Growing evidence implicates the gut microbiota in balancing and modulating immunity response, and in the pathogenesis of allergic disease. As a result, gut microbial imbalance could be a useful indicator for allergic conjunctivitis. From the perspective of predictive, preventive, and personalized medicine (PPPM), clarifying the role of gut microbial imbalance in the development of allergic conjunctivitis could provide a window of opportunity for primary prediction, targeted prevention, and personalized treatment of the disease. Working hypothesis and methodology In our study, we hypothesized that individuals with microbial dysbiosis may be more susceptible to allergic conjunctivitis due to an increased inflammatory response. To verify the working hypothesis, our analysis selected genetic variants linked with gut microbiota features (N = 18,340) and allergic conjunctivitis (4513 cases, 649,376 controls) from genome-wide association studies. The inverse-variance weighted (IVW) estimate, Mendelian randomization (MR)-Egger, weighted median estimator, maximum likelihood estimator (MLE), and MR robust adjusted profile score (MR.RAPS) were employed to analyze the impact of gut microbiota on the risk of allergic conjunctivitis and identify allergic conjunctivitis-related gut microbes. Ultimately, these findings may enable the identification of individuals at risk of allergic conjunctivitis through screening of gut microbial imbalances, and allow for new targeted prevention and personalized treatment strategies. Results Genetic liability to Ruminococcaceae_UCG_002 (OR, 0.83; 95% CI, 0.70-0.99; P = 4.04×10-2), Holdemanella (OR, 0.78; 95% CI, 0.64-0.96; P = 2.04×10-2), Catenibacterium (OR, 0.69; 95% CI, 0.56-0.86; P = 1.09×10-3), Senegalimassilia (OR, 0.71; 95% CI, 0.55-0.93; P = 1.23×10-2) genus were associated with a low risk of allergic conjunctivitis with IVW. Besides, we found suggestive associations of a genetic-driven increase in the Oscillospira (OR, 1.41; 95% CI, 1.00-2.00; P = 4.63×10-2) genus with a higher risk of allergic conjunctivitis. Moreover, MLE and MR.RAPS show consistent results with IVW after further validation and strengthened confidence in the true causal associations. No heterogeneity and pleiotropy was detected. Conclusions Our study suggests that gut microbiota may play a causal role in the development of allergic conjunctivitis and provides new insights into the microbiota-mediated mechanism of the disease. Gut microbiota may serve as a target for future predictive diagnostics, targeted prevention, and individualized therapy in allergic conjunctivitis, facilitating the transition from reactive medical services to PPPM in the management of the disease. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-023-00321-9.
Collapse
Affiliation(s)
- Kangcheng Liu
- No.87, Xiangya Road, Kaifu District, Changsha, 410008 Hunan Province China Eye Center of Xiangya Hospital, Central South University
- Changsha, 410008 Hunan Province China Hunan Key Laboratory of Ophthalmology
- Changsha, 410008 Hunan Province China National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University
- Nanchang, China Jiangxi Province Division of National Clinical Research Center for Ocular Diseases, Jiangxi Clinical Research Center for Ophthalmic Disease, Jiangxi Research Institute of Ophthalmology and Visual Science, Affiliated Eye Hospital of Nanchang University
| | - Yingjun Cai
- No.87, Xiangya Road, Kaifu District, Changsha, 410008 Hunan Province China Eye Center of Xiangya Hospital, Central South University
- Changsha, 410008 Hunan Province China Hunan Key Laboratory of Ophthalmology
- Changsha, 410008 Hunan Province China National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University
| | - Kun Song
- Changsha, 410008 Hunan Province China Department of Gastrointestinal Surgery, Xiangya Hospital, Central South University
| | - Ruolan Yuan
- No.87, Xiangya Road, Kaifu District, Changsha, 410008 Hunan Province China Eye Center of Xiangya Hospital, Central South University
- Changsha, 410008 Hunan Province China Hunan Key Laboratory of Ophthalmology
- Changsha, 410008 Hunan Province China National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University
| | - Jing Zou
- No.87, Xiangya Road, Kaifu District, Changsha, 410008 Hunan Province China Eye Center of Xiangya Hospital, Central South University
- Changsha, 410008 Hunan Province China Hunan Key Laboratory of Ophthalmology
- Changsha, 410008 Hunan Province China National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University
| |
Collapse
|
19
|
Wu Y, Zhang G, Wang Y, Wei X, Liu H, Zhang L, Zhang L. A Review on Maternal and Infant Microbiota and Their Implications for the Prevention and Treatment of Allergic Diseases. Nutrients 2023; 15:nu15112483. [PMID: 37299446 DOI: 10.3390/nu15112483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 06/12/2023] Open
Abstract
Allergic diseases, which are closely related to the composition and metabolism of maternal and infant flora, are prevalent in infants worldwide. The mother's breast milk, intestinal, and vaginal flora directly or indirectly influence the development of the infant's immune system from pregnancy to lactation, and the compositional and functional alterations of maternal flora are associated with allergic diseases in infants. Meanwhile, the infant's own flora, represented by the intestinal flora, indicates and regulates the occurrence of allergic diseases and is altered with the intervention of allergic diseases. By searching and selecting relevant literature in PubMed from 2010 to 2023, the mechanisms of allergy development in infants and the links between maternal and infant flora and infant allergic diseases are reviewed, including the effects of flora composition and its consequences on infant metabolism. The critical role of maternal and infant flora in allergic diseases has provided a window for probiotics as a microbial therapy. Therefore, the uses and mechanisms by which probiotics, such as lactic acid bacteria, can help to improve the homeostasis of both the mother and the infant, and thereby treat allergies, are also described.
Collapse
Affiliation(s)
- Yifan Wu
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Gongsheng Zhang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Yucong Wang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Xin Wei
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Huanhuan Liu
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Lili Zhang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Lanwei Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266100, China
| |
Collapse
|
20
|
Liang Y, Xie R, Xiong X, Hu Z, Mao X, Wang X, Zhang J, Sun P, Yue Z, Wang W, Zhang G. Alterations of nasal microbiome in eosinophilic chronic rhinosinusitis. J Allergy Clin Immunol 2023; 151:1286-1295.e2. [PMID: 36736796 DOI: 10.1016/j.jaci.2022.11.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 10/16/2022] [Accepted: 11/04/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND Exposure to microbes may be important in the development of chronic rhinosinusitis (CRS). Dysbiosis of the nasal microbiome is considered to be related to CRS with nasal polyps (CRSwNP). The link between the nasal microbiota and eosinophilic CRSwNP (eCRSwNP) has rarely been studied. OBJECTIVE The aim of this study was to rigorously characterize nasal dysbiosis in a cohort of patients with eCRSwNP and compare the nasal microbiomes of these patients with those of healthy controls (HCs). METHODS We performed a cross-sectional study of 34 patients with eCRSwNP, 10 patients without CRSwNP, and 44 HCs by using 16S rRNA gene sequencing. An independent cohort of 14 patients with eCRSwNP, 9 patients without CRSwNP, and 11 HCs was used to validate the results. RESULTS Compared with the nasal microbiome of healthy controls, the nasal microbiome of patients with eCRSwNP was characterized by higher α-diversity (Shannon and Chao1 index) and a distinct composition of microbes. Notably, the distinct differences in microbial composition between patients with eCRSwNP and HCs were significantly correlated with eCRSwNP disease status. Furthermore, in a diagnostic model generated by using these differences, a combination of 15 genera could be used to distinguish patients with eCRSwNP from HCs, with an area under the curve of approximately 0.8 in both the exploration and validation cohorts. CONCLUSION Our study establishes the compositional alterations in the nasal microbiome in eCRSwNP and suggests the potential for using the nasal microbiota as a noninvasive predictive classifier for the diagnosis of eCRSwNP.
Collapse
Affiliation(s)
- Yibo Liang
- Department of Otorhinolaryngology Head and Neck Surgery, Tianjin First Central Hospital, Tianjin, China; Institute of Otolaryngology of Tianjin, Tianjin, China; Key Laboratory of Auditory Speech and Balance Medicine, Tianjin; Key Medical Discipline of Tianjin (Otolaryngology), Tianjin, China; Quality Control Centre of Otolaryngology, Tianjin, China
| | - Runxiang Xie
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Xin Xiong
- Department of Otorhinolaryngology Head and Neck Surgery, Tianjin First Central Hospital, Tianjin, China; Institute of Otolaryngology of Tianjin, Tianjin, China; Key Laboratory of Auditory Speech and Balance Medicine, Tianjin; Key Medical Discipline of Tianjin (Otolaryngology), Tianjin, China; Quality Control Centre of Otolaryngology, Tianjin, China
| | - Zhanjun Hu
- Department of Pathology, Tianjin First Central Hospital, Tianjin, China
| | - Xiang Mao
- Department of Otorhinolaryngology Head and Neck Surgery, Tianjin First Central Hospital, Tianjin, China; Institute of Otolaryngology of Tianjin, Tianjin, China; Key Laboratory of Auditory Speech and Balance Medicine, Tianjin; Key Medical Discipline of Tianjin (Otolaryngology), Tianjin, China; Quality Control Centre of Otolaryngology, Tianjin, China
| | - Xiaoyu Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Tianjin First Central Hospital, Tianjin, China; Institute of Otolaryngology of Tianjin, Tianjin, China; Key Laboratory of Auditory Speech and Balance Medicine, Tianjin; Key Medical Discipline of Tianjin (Otolaryngology), Tianjin, China; Quality Control Centre of Otolaryngology, Tianjin, China
| | - Jinmei Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Tianjin First Central Hospital, Tianjin, China; Institute of Otolaryngology of Tianjin, Tianjin, China; Key Laboratory of Auditory Speech and Balance Medicine, Tianjin; Key Medical Discipline of Tianjin (Otolaryngology), Tianjin, China; Quality Control Centre of Otolaryngology, Tianjin, China
| | - Peiyong Sun
- Department of Otorhinolaryngology Head and Neck Surgery, Tianjin First Central Hospital, Tianjin, China; Institute of Otolaryngology of Tianjin, Tianjin, China; Key Laboratory of Auditory Speech and Balance Medicine, Tianjin; Key Medical Discipline of Tianjin (Otolaryngology), Tianjin, China; Quality Control Centre of Otolaryngology, Tianjin, China
| | - Zhenzhong Yue
- Department of Otorhinolaryngology Head and Neck Surgery, Tianjin First Central Hospital, Tianjin, China; Institute of Otolaryngology of Tianjin, Tianjin, China; Key Laboratory of Auditory Speech and Balance Medicine, Tianjin; Key Medical Discipline of Tianjin (Otolaryngology), Tianjin, China; Quality Control Centre of Otolaryngology, Tianjin, China
| | - Wei Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Tianjin First Central Hospital, Tianjin, China; Institute of Otolaryngology of Tianjin, Tianjin, China; Key Laboratory of Auditory Speech and Balance Medicine, Tianjin; Key Medical Discipline of Tianjin (Otolaryngology), Tianjin, China; Quality Control Centre of Otolaryngology, Tianjin, China
| | - Guimin Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Tianjin First Central Hospital, Tianjin, China; Institute of Otolaryngology of Tianjin, Tianjin, China; Key Laboratory of Auditory Speech and Balance Medicine, Tianjin; Key Medical Discipline of Tianjin (Otolaryngology), Tianjin, China; Quality Control Centre of Otolaryngology, Tianjin, China.
| |
Collapse
|
21
|
Kesavelu D, Jog P. Current understanding of antibiotic-associated dysbiosis and approaches for its management. Ther Adv Infect Dis 2023; 10:20499361231154443. [PMID: 36860273 PMCID: PMC9969474 DOI: 10.1177/20499361231154443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 01/16/2023] [Indexed: 03/03/2023] Open
Abstract
Increased exposure to antibiotics during early childhood increases the risk of antibiotic-associated dysbiosis, which is associated with reduced diversity of gut microbial species and abundance of certain taxa, disruption of host immunity, and the emergence of antibiotic-resistant microbes. The disruption of gut microbiota and host immunity in early life is linked to the development of immune-related and metabolic disorders later in life. Antibiotic administration in populations predisposed to gut microbiota dysbiosis, such as newborns, obese children, and children with allergic rhinitis and recurrent infections; changes microbial composition and diversity; exacerbating dysbiosis and resulting in negative health outcomes. Antibiotic-associated diarrhea (AAD), Clostridiodes difficile-associated diarrhea (CDAD), and Helicobacter pylori infection are all short-term consequences of antibiotic treatment that persist from a few weeks to months. Changes in gut microbiota, which persist even 2 years after antibiotic exposure, and the development of obesity, allergies, and asthma are among the long-term consequences. Probiotic bacteria and dietary supplements can potentially prevent or reverse antibiotic-associated gut microbiota dysbiosis. Probiotics have been demonstrated in clinical studies to help prevent AAD and, to a lesser extent, CDAD, as well as to improve H pylori eradication rates. In the Indian setting, probiotics (Saccharomyces boulardii and Bacillus clausii) have been shown to reduce the duration and frequency of acute diarrhea in children. Antibiotics may exaggerate the consequences of gut microbiota dysbiosis in vulnerable populations already affected by the condition. Therefore, prudent use of antibiotics among neonates and young children is critical to prevent the detrimental effects on gut health.
Collapse
Affiliation(s)
| | - Pramod Jog
- Dr. D.Y. Patil Medical College, Hospital &
Research Centre, Pune, India
| |
Collapse
|
22
|
Sjödin KS, Sjödin A, Ruszczyński M, Kristensen MB, Hernell O, Szajewska H, West CE. Targeting the gut-lung axis by synbiotic feeding to infants in a randomized controlled trial. BMC Biol 2023; 21:38. [PMID: 36803508 PMCID: PMC9940374 DOI: 10.1186/s12915-023-01531-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 01/27/2023] [Indexed: 02/21/2023] Open
Abstract
BACKGROUND Formula-fed infants are at increased risk of infections. Due to the cross-talk between the mucosal systems of the gastrointestinal and respiratory tracts, adding synbiotics (prebiotics and probiotics) to infant formula may prevent infections even at distant sites. Infants that were born full term and weaned from breast milk were randomized to prebiotic formula (fructo- and galactooligosaccharides) or the same prebiotic formula with Lactobacillus paracasei ssp. paracasei F19 (synbiotics) from 1 to 6 months of age. The objective was to examine the synbiotic effects on gut microbiota development. RESULTS Fecal samples collected at ages 1, 4, 6, and 12 months were analyzed using 16S rRNA gene sequencing and a combination of untargeted gas chromatography-mass spectrometry/liquid chromatography-mass spectrometry. These analyses revealed that the synbiotic group had a lower abundance of Klebsiella, a higher abundance of Bifidobacterium breve compared to the prebiotic group, and increases in the anti-microbial metabolite d-3-phenyllactic acid. We also analyzed the fecal metagenome and antibiotic resistome in the 11 infants that had been diagnosed with lower respiratory tract infection (cases) and 11 matched controls using deep metagenomic sequencing. Cases with lower respiratory tract infection had a higher abundance of Klebsiella species and antimicrobial resistance genes related to Klebsiella pneumoniae, compared to controls. The results obtained using 16S rRNA gene amplicon and metagenomic sequencing were confirmed in silico by successful recovery of the metagenome-assembled genomes of the bacteria of interest. CONCLUSIONS This study demonstrates the additional benefit of feeding specific synbiotics to formula-fed infants over prebiotics only. Synbiotic feeding led to the underrepresentation of Klebsiella, enrichment of bifidobacteria, and increases in microbial degradation metabolites implicated in immune signaling and in the gut-lung and gut-skin axes. Our findings support future clinical evaluation of synbiotic formula in the prevention of infections and associated antibiotic treatment as a primary outcome when breastfeeding is not feasible. TRIAL REGISTRATION ClinicalTrials.gov NCT01625273 . Retrospectively registered on 21 June 2012.
Collapse
Affiliation(s)
- Kotryna Simonyté Sjödin
- grid.12650.300000 0001 1034 3451Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, 901 85 Sweden
| | - Andreas Sjödin
- grid.12650.300000 0001 1034 3451Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, 901 85 Sweden ,Division of CBRN Security and Defense, FOI – Swedish Defense Research Agency, Umeå, Sweden
| | - Marek Ruszczyński
- grid.13339.3b0000000113287408Department of Paediatrics, The Medical University of Warsaw, Warsaw, Poland
| | | | - Olle Hernell
- grid.12650.300000 0001 1034 3451Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, 901 85 Sweden
| | - Hania Szajewska
- grid.13339.3b0000000113287408Department of Paediatrics, The Medical University of Warsaw, Warsaw, Poland
| | - Christina E. West
- grid.12650.300000 0001 1034 3451Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, 901 85 Sweden
| |
Collapse
|
23
|
Lif Holgerson P, Esberg A, West CE, Johansson I. The breast milk and childhood gastrointestinal microbiotas and disease outcomes: a longitudinal study. Pediatr Res 2023; 93:570-578. [PMID: 36216869 PMCID: PMC9988688 DOI: 10.1038/s41390-022-02328-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 09/10/2022] [Accepted: 09/14/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND We aimed to characterize breast milk microbiota and define associations with saliva and fecal microbiota and selected diseases in preschool children. METHODS In a longitudinal cohort study, the microbiotas from breast milk, mouth, and fecal samples were characterized by 16S rRNA gene sequencing. Questionnaires and medical records provided information on demographics, medical, and dental data. RESULTS The phylogeny in breast milk, saliva swabs, and feces differed at all levels (p < 0.0003), though all harbored species in Streptococcus, Veillonella, and Haemophilus. Species richness was highest in breast milk with increasing resemblance with the oral swab microbiota by increasing age. Caries-affected children at age 5 had been fed breast milk with tenfold higher abundance of caries-associated bacteria, e.g., Streptococcus mutans, than caries-free children (p < 0.002). At that age, taxa, e.g., Neisseria sicca were overrepresented in saliva swabs of children with otitis media (LDA score >2, p < 0.05). Gut symbionts, e.g., Bacteroides, were underrepresented in 3-month fecal samples in children later diagnosed with allergic disease (LDA score >2, p < 0.05). CONCLUSIONS Distinct microbiotas for the three sources were confirmed, though resemblance between milk and oral swab microbiota increased by age. Future studies should evaluate if the observed associations with disease outcomes are causal. IMPACT Few studies have studied the association between breast milk microbiota and gastrointestinal microbiota beyond early infancy. The present study confirms distinct microbiota profiles in breast milk, saliva swabs, and feces in infancy and indicates increasing resemblance between breast milk and the oral microbiota by increasing age. The fecal microbiota at 3 months was associated with later allergic disease; the saliva microbiota by age 5 differed between children with and without otitis media at the same age; and children with caries by age 5 had been fed breast milk with a higher abundance of caries-associated bacteria.
Collapse
Affiliation(s)
- Pernilla Lif Holgerson
- Department of Odontology, Section of Pediatric Dentistry, Umeå University, Umeå, Sweden.
| | - Anders Esberg
- Department of Odontology, Section of Cariology, Umeå University, Umeå, Sweden
| | - Christina E West
- Department of Clinical Sciences, Section of Pediatrics, Umeå University, Umeå, Sweden
| | - Ingegerd Johansson
- Department of Odontology, Section of Cariology, Umeå University, Umeå, Sweden
| |
Collapse
|
24
|
Sadeghpour Heravi F, Hu H. Bifidobacterium: Host-Microbiome Interaction and Mechanism of Action in Preventing Common Gut-Microbiota-Associated Complications in Preterm Infants: A Narrative Review. Nutrients 2023; 15:709. [PMID: 36771414 PMCID: PMC9919561 DOI: 10.3390/nu15030709] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 01/23/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
The development and health of infants are intertwined with the protective and regulatory functions of different microorganisms in the gut known as the gut microbiota. Preterm infants born with an imbalanced gut microbiota are at substantial risk of several diseases including inflammatory intestinal diseases, necrotizing enterocolitis, late-onset sepsis, neurodevelopmental disorders, and allergies which can potentially persist throughout adulthood. In this review, we have evaluated the role of Bifidobacterium as commonly used probiotics in the development of gut microbiota and prevention of common diseases in preterm infants which is not fully understood yet. The application of Bifidobacterium as a therapeutical approach in the re-programming of the gut microbiota in preterm infants, the mechanisms of host-microbiome interaction, and the mechanism of action of this bacterium have also been investigated, aiming to provide new insights and opportunities in microbiome-targeted interventions in personalized medicine.
Collapse
Affiliation(s)
| | - Honghua Hu
- Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia
- Innovation Center of Translational Pharmacy, Jinhua Institute of Zhejiang University, Jinhua 321016, China
| |
Collapse
|
25
|
Yamaguchi T, Nomura A, Matsubara A, Hisada T, Tamada Y, Mikami T, Ishida M. Effect of gut microbial composition and diversity on major inhaled allergen sensitization and onset of allergic rhinitis. Allergol Int 2023; 72:135-142. [PMID: 35850746 DOI: 10.1016/j.alit.2022.06.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/24/2022] [Accepted: 06/10/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Decreased gut microbiota diversity is associated with gut dysbiosis and causes various diseases, including allergic diseases. We investigated the relationship between gut microbial diversity and sensitization to major inhaled allergens. Furthermore, the relationship of allergic symptom onset with bacterial composition in sensitized individuals was investigated. METHODS This study included 1092 local residents who had participated in the Iwaki Health Promotion Project in 2016. Blood samples were analyzed to ascertain specific IgE levels against major inhaled allergens (JCP, HD1, Grass-mix, Weed-mix). Nasal symptoms were estimated by questionnaires. Fecal samples were analyzed for bacterial 16S rRNA using next generation sequencing. The diversity index (α-diversity, β-diversity) and the composition of gut microbes in phylum/order levels were compared between patients sensitized or unsensitized to allergen, and symptomatic and asymptomatic groups. RESULTS Some α-diversity metrics were significantly decreased in patients who were sensitized to any/all four allergens compared with the unsensitized group. β-diversity differed significantly between those unsensitized and sensitized to all allergens (aged 20-49 years), and between those unsensitized and sensitized to any/all four allergens (aged ≥50 years). The relative abundance of Bacteroidales was significantly lower in the unsensitized than in the sensitized group. The composition and diversity of gut microbiota were similar between the symptomatic and asymptomatic groups. CONCLUSIONS Our results suggest that lack of diversity in gut microbiota has an effect on sensitization to allergens. Bacteroidales in order level may affect sensitization; however, the onset of allergy symptoms was not significantly associated with bacterial composition and diversity.
Collapse
Affiliation(s)
- Taimu Yamaguchi
- Department of Otorhinolaryngology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Ayami Nomura
- Department of Otorhinolaryngology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Atsushi Matsubara
- Department of Otorhinolaryngology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan.
| | | | - Yoshinori Tamada
- Department of Social Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Tatsuya Mikami
- Department of Social Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Mizuri Ishida
- Department of Social Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| |
Collapse
|
26
|
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
|
27
|
Worm M, Alexiou A, Höfer V, Birkner T, Jeanrenaud ACSN, Fauchère F, Pazur K, Steinert C, Arnau‐Soler A, Banerjee P, Diefenbach A, Dobbertin‐Welsch J, Dölle‐Bierke S, Francuzik W, Ghauri A, Heller S, Kalb B, Löber U, Marenholz I, Markó L, Scheffel J, Potapenko O, Roll S, Lau S, Lee Y, Braun J, Thiel A, Babina M, Altrichter S, Forslund SK, Beyer K. An interdisciplinary approach to characterize peanut-allergic patients-First data from the FOOD@ consortium. Clin Transl Allergy 2022; 12:e12197. [PMID: 36225266 PMCID: PMC9533219 DOI: 10.1002/clt2.12197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/29/2022] [Accepted: 09/05/2022] [Indexed: 12/01/2022] Open
Abstract
Background Peanut allergy is a frequent cause of food allergy and potentially life-threatening. Within this interdisciplinary research approach, we aim to unravel the complex mechanisms of peanut allergy. As a first step were applied in an exploratory manner the analysis of peanut allergic versus non-allergic controls. Methods Biosamples were studied regarding DNA methylation signatures, gut microbiome, adaptive and innate immune cell populations, soluble signaling molecules and allergen-reactive antibody specificities. We applied a scalable systems medicine computational workflow to the assembled data. Results We identified combined cellular and soluble biomarker signatures that stratify donors into peanut-allergic and non-allergic with high specificity. DNA methylation profiling revealed various genes of interest and stool microbiota differences in bacteria abundances. Conclusion By extending our findings to a larger set of patients (e.g., children vs. adults), we will establish predictors for food allergy and tolerance and translate these as for example, indicators for interventional studies.
Collapse
Affiliation(s)
- Margitta Worm
- Division of Allergy and ImmunologyDepartment of Dermatology, Venerology and AllergyCharité – Universitätsmedizin BerlinFreie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- KFO339, FOOD@BerlinGermany
| | - Aikaterina Alexiou
- Division of Allergy and ImmunologyDepartment of Dermatology, Venerology and AllergyCharité – Universitätsmedizin BerlinFreie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- KFO339, FOOD@BerlinGermany
| | - Veronika Höfer
- Division of Allergy and ImmunologyDepartment of Dermatology, Venerology and AllergyCharité – Universitätsmedizin BerlinFreie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- KFO339, FOOD@BerlinGermany
| | - Till Birkner
- Experimental and Clinical Research CenterA Cooperation of Charité‐Universitätsmedizin BerlinMax Delbrück Center for Molecular MedicineBerlinGermany
- Charité‐Universitätsmedizin BerlinFreie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of HealthBerlinGermany
- Max Delbrück Center for Molecular MedicineHelmholtz AssociationBerlinGermany
- KFO339, FOOD@BerlinGermany
| | - Alexander C. S. N. Jeanrenaud
- Max Delbrück Center for Molecular MedicineHelmholtz AssociationBerlinGermany
- Clinic for Pediatric Allergy, Experimental and Clinical Research CenterCharité – Universitätsmedizin BerlinFreie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- KFO339, FOOD@BerlinGermany
| | - Florent Fauchère
- Si‐M/“Der Simulierte Mensch” a Science Framework of Technische Universität Berlin and Charité – Universitätsmedizin BerlinBerlinGermany
- Regenerative Immunology and AgingBIH Center for Regenerative TherapiesCharité ‐ Universitätsmedizin BerlinCorporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- KFO339, FOOD@BerlinGermany
| | - Kristijan Pazur
- Division of Allergy and ImmunologyDepartment of Dermatology, Venerology and AllergyCharité – Universitätsmedizin BerlinFreie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- KFO339, FOOD@BerlinGermany
| | - Carolin Steinert
- Institute of Allergology IFACharité‐Universitätsmedizin BerlinFreie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of HealthBerlinGermany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMPAllergology and Immunology AIBerlinGermany
- Department of Biology, Chemistry and PharmacyFreie Universität BerlinBerlinGermany
- KFO339, FOOD@BerlinGermany
| | - Aleix Arnau‐Soler
- Max Delbrück Center for Molecular MedicineHelmholtz AssociationBerlinGermany
- Clinic for Pediatric Allergy, Experimental and Clinical Research CenterCharité – Universitätsmedizin BerlinFreie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- KFO339, FOOD@BerlinGermany
| | - Priyanka Banerjee
- Institute of PhysiologyCharité – Universitätsmedizin BerlinFreie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of HealthBerlinGermany
- KFO339, FOOD@BerlinGermany
| | - Andreas Diefenbach
- Mucosal and Developmental ImmunologyGerman Rheuma Research Center Berlin (DRFZ)BerlinGermany
- Department of Microbiology, Infectious Diseases, and ImmunologyLaboratory of Innate ImmunityCharité – Universitätsmedizin BerlinCampus Benjamin FranklinBerlinGermany
- KFO339, FOOD@BerlinGermany
| | - Josefine Dobbertin‐Welsch
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care MedicineCharité – Universitätsmedizin BerlinFreie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- KFO339, FOOD@BerlinGermany
| | - Sabine Dölle‐Bierke
- Division of Allergy and ImmunologyDepartment of Dermatology, Venerology and AllergyCharité – Universitätsmedizin BerlinFreie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- KFO339, FOOD@BerlinGermany
| | - Wojciech Francuzik
- Division of Allergy and ImmunologyDepartment of Dermatology, Venerology and AllergyCharité – Universitätsmedizin BerlinFreie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- KFO339, FOOD@BerlinGermany
| | - Ahla Ghauri
- Max Delbrück Center for Molecular MedicineHelmholtz AssociationBerlinGermany
- Clinic for Pediatric Allergy, Experimental and Clinical Research CenterCharité – Universitätsmedizin BerlinFreie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- KFO339, FOOD@BerlinGermany
| | - Stephanie Heller
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care MedicineCharité – Universitätsmedizin BerlinFreie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- KFO339, FOOD@BerlinGermany
| | - Birgit Kalb
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care MedicineCharité – Universitätsmedizin BerlinFreie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- KFO339, FOOD@BerlinGermany
| | - Ulrike Löber
- Experimental and Clinical Research CenterA Cooperation of Charité‐Universitätsmedizin BerlinMax Delbrück Center for Molecular MedicineBerlinGermany
- Charité‐Universitätsmedizin BerlinFreie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of HealthBerlinGermany
- Max Delbrück Center for Molecular MedicineHelmholtz AssociationBerlinGermany
- KFO339, FOOD@BerlinGermany
| | - Ingo Marenholz
- Max Delbrück Center for Molecular MedicineHelmholtz AssociationBerlinGermany
- Clinic for Pediatric Allergy, Experimental and Clinical Research CenterCharité – Universitätsmedizin BerlinFreie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- KFO339, FOOD@BerlinGermany
| | - Lajos Markó
- Experimental and Clinical Research CenterA Cooperation of Charité‐Universitätsmedizin BerlinMax Delbrück Center for Molecular MedicineBerlinGermany
- Charité‐Universitätsmedizin BerlinFreie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of HealthBerlinGermany
- Max Delbrück Center for Molecular MedicineHelmholtz AssociationBerlinGermany
- KFO339, FOOD@BerlinGermany
| | - Jörg Scheffel
- Institute of Allergology IFACharité‐Universitätsmedizin BerlinFreie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of HealthBerlinGermany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMPAllergology and Immunology AIBerlinGermany
- KFO339, FOOD@BerlinGermany
| | - Olena Potapenko
- Experimental and Clinical Research CenterA Cooperation of Charité‐Universitätsmedizin BerlinMax Delbrück Center for Molecular MedicineBerlinGermany
- Charité‐Universitätsmedizin BerlinFreie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of HealthBerlinGermany
- Max Delbrück Center for Molecular MedicineHelmholtz AssociationBerlinGermany
- KFO339, FOOD@BerlinGermany
| | - Stephanie Roll
- Institute of Social Medicine, Epidemiology and Health EconomicsCharité—Universitätsmedizin BerlinFreie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- KFO339, FOOD@BerlinGermany
| | - Susanne Lau
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care MedicineCharité – Universitätsmedizin BerlinFreie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- KFO339, FOOD@BerlinGermany
| | - Young‐Ae Lee
- Max Delbrück Center for Molecular MedicineHelmholtz AssociationBerlinGermany
- Clinic for Pediatric Allergy, Experimental and Clinical Research CenterCharité – Universitätsmedizin BerlinFreie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- KFO339, FOOD@BerlinGermany
| | - Julian Braun
- Si‐M/“Der Simulierte Mensch” a Science Framework of Technische Universität Berlin and Charité – Universitätsmedizin BerlinBerlinGermany
- Regenerative Immunology and AgingBIH Center for Regenerative TherapiesCharité ‐ Universitätsmedizin BerlinCorporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- KFO339, FOOD@BerlinGermany
| | - Andreas Thiel
- Si‐M/“Der Simulierte Mensch” a Science Framework of Technische Universität Berlin and Charité – Universitätsmedizin BerlinBerlinGermany
- Regenerative Immunology and AgingBIH Center for Regenerative TherapiesCharité ‐ Universitätsmedizin BerlinCorporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- KFO339, FOOD@BerlinGermany
| | - Magda Babina
- Institute of Allergology IFACharité‐Universitätsmedizin BerlinFreie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of HealthBerlinGermany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMPAllergology and Immunology AIBerlinGermany
- KFO339, FOOD@BerlinGermany
| | - Sabine Altrichter
- Institute of Allergology IFACharité‐Universitätsmedizin BerlinFreie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of HealthBerlinGermany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMPAllergology and Immunology AIBerlinGermany
- Department of Dermatology and VenerologyKepler University HospitalLinzAustria
- KFO339, FOOD@BerlinGermany
| | - Sofia Kirke Forslund
- Experimental and Clinical Research CenterA Cooperation of Charité‐Universitätsmedizin BerlinMax Delbrück Center for Molecular MedicineBerlinGermany
- Charité‐Universitätsmedizin BerlinFreie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of HealthBerlinGermany
- Max Delbrück Center for Molecular MedicineHelmholtz AssociationBerlinGermany
- KFO339, FOOD@BerlinGermany
| | - Kirsten Beyer
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care MedicineCharité – Universitätsmedizin BerlinFreie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- KFO339, FOOD@BerlinGermany
| |
Collapse
|
28
|
Kelderer F, Mogren I, Eriksson C, Silfverdal SA, Domellöf M, West CE. Associations between pre- and postnatal antibiotic exposures and early allergic outcomes: A population-based birth cohort study. Pediatr Allergy Immunol 2022; 33:e13848. [PMID: 36156813 PMCID: PMC9544930 DOI: 10.1111/pai.13848] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 07/21/2022] [Accepted: 08/16/2022] [Indexed: 12/03/2022]
Abstract
BACKGROUND Early life antibiotic treatment is one likely exposure influencing allergy risk. The objective was to investigate associations between pre- and postnatal antibiotic exposures and the development of allergic manifestations until age 18 months. METHODS We included 1387 mother-child dyads from the prospective, population-based NorthPop birth cohort study. Data on antibiotic exposures in pregnancy and childhood were collected by web-based questionnaires. Until the child turned 18 months old, parents (n = 1219) reported symptoms of wheeze, eczema, and physician-diagnosed asthma; parents (n = 1025) reported physician-diagnosed food allergy. At age 18 months, serum immunoglobulin E levels to inhalant (Phadiatop) and food (Food mix fx5) allergens were determined. Associations were estimated using bivariable and multivariable logistic regressions. RESULTS Prenatal antibiotic exposure was positively associated with food sensitization in the crude (OR 1.82, 95% CI 1.01-3.26) but not in the adjusted analyses (aOR 1.58, 0.82-3.05). A borderline significant association was found between prenatal exposure and wheeze (aOR 1.56, 0.95-2.57). Postnatal antibiotics were positively associated with wheeze (aOR 2.14, 1.47-3.11), asthma (aOR 2.35, 1.32-4.19), and eczema (aOR 1.49, 1.07-2.06). Postnatal antibiotics were negatively associated with food sensitization (aOR 0.46, 95% CI 0.25-0.83) but not with food allergy nor sensitization to inhalants. CONCLUSION Pre- and postnatal antibiotic exposure demonstrated positive associations with allergic manifestations and the former also with food sensitization. In contrast, there was a negative association between postnatal antibiotics and food sensitization. Food sensitization is often transient but may precede respiratory allergies. Future studies should investigate the relationship between antibiotic exposure and food sensitization later in childhood.
Collapse
Affiliation(s)
- Fanny Kelderer
- Department of Clinical Sciences, Pediatrics Umeå University, Umeå, Sweden
| | - Ingrid Mogren
- Department of Clinical Sciences, Obstetrics and Gynecology, Umeå, Sweden
| | - Catharina Eriksson
- Department of Clinical Microbiology, Infection and Immunology, Umeå University, Umeå, Sweden
| | | | - Magnus Domellöf
- Department of Clinical Sciences, Pediatrics Umeå University, Umeå, Sweden
| | - Christina E West
- Department of Clinical Sciences, Pediatrics Umeå University, Umeå, Sweden
| |
Collapse
|
29
|
Liao M, Xiao Y, Li S, Su J, Li J, Zou B, Chen X, Shen M. Synergistic Effects between Ambient Air Pollution and Second-Hand Smoke on Inflammatory Skin Diseases in Chinese Adolescents. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:10011. [PMID: 36011645 PMCID: PMC9408277 DOI: 10.3390/ijerph191610011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/14/2022] [Accepted: 08/05/2022] [Indexed: 06/15/2023]
Abstract
Atopic dermatitis (AD), chronic hand eczema (CHE), and urticaria are common inflammatory skin diseases among adolescents and associated with air quality. However, the synergistic effects of ambient air pollution and second-hand smoke (SHS) have been unclear. We conducted a cross-sectional study including 20,138 Chinese college students where dermatological examinations and a questionnaire survey were carried out. A generalized linear mixed model was applied for the association between individualized exposure of O3, CO, NO2, SO2, PM2.5, and PM10 and the prevalence of inflammatory skin diseases. Interactions between air pollutants and SHS were analyzed. As a result, CO, NO2, SO2, PM2.5, and PM10 were positively correlated with the prevalence of AD, CHE, and urticaria. Higher frequency of SHS exposure contributed to increased probabilities of AD (p = 0.042), CHE (p < 0.001), and urticaria (p = 0.002). Of note, CO (OR: 2.57 (1.16−5.69) in third quartile) and NO2 (OR: 2.38 (1.07−5.27) in third quartile) had positive interactions with SHS for AD, and PM2.5 synergized with SHS for CHE (OR: 2.25 (1.22−4.15) for second quartile). Subgroup analyses agreed with the synergistic results. In conclusion, SHS and ambient air pollution are both associated with inflammatory skin diseases, and they have a synergistic effect on the prevalence of AD and CHE.
Collapse
Affiliation(s)
- Mengting Liao
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, China
- Health Management Center, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yi Xiao
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, China
| | - Shenxin Li
- Department of Surveying and Remote Sensing Science, School of Geosciences and Info-Physics, Central South University, Changsha 410008, China
| | - Juan Su
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ji Li
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Bin Zou
- Department of Surveying and Remote Sensing Science, School of Geosciences and Info-Physics, Central South University, Changsha 410008, China
| | - Xiang Chen
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Minxue Shen
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Social Medicine and Health Management, Xiangya School of Public Health, Central South University, Changsha 410008, China
| |
Collapse
|
30
|
林 一, 李 俊, 李 幼. [Metagenome-wide association of gut microbiome features in children with moderate-severe house dust mite allergic rhinitis]. LIN CHUANG ER BI YAN HOU TOU JING WAI KE ZA ZHI = JOURNAL OF CLINICAL OTORHINOLARYNGOLOGY, HEAD, AND NECK SURGERY 2022; 36:533-539. [PMID: 35822382 PMCID: PMC10128392 DOI: 10.13201/j.issn.2096-7993.2022.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Indexed: 06/15/2023]
Abstract
Objective:To draw a distinct gut microbiota pattern of children with moderate-severe dust mite-induced allergic rhinitis(DAR) and healthy children. Methods:3-10 years old moderate-severe DAR children(68 cases) and healthy children(38 cases) were involved in this study. General information was collected through questionnaires, and fecal samples were collected for metagenomic sequencing. MetaPhlAn3 was used to generate the microbiota composition abundance in detail, and Alpha and Beta diversity changes were calculated. The difference in species abundance at different taxonomic levels were compared. Differences in functional pathways were compared by LEfSe analysis. Results:The diversity of gut microbiota in children with moderate-severe DAR didn't change significantly compared with healthy children. A total of 37 microbial communities or species with significant abundance difference were found, mainly included Lachnoclostridium, Prevotella, Blautia wexlerae, Prevotella copri, Eubacterium eligens, Eubacterium sp CAG 180, etc. However, the metabolism functions of gut microbiota in children with moderate-severe DAR changed compared with healthy children. Various of fatty acids anabolism enhanced in DAR children. Conclusion:Compared with healthy children, there was no significant difference in gut microbial diversity in moderate-severe DAR children. The abundance of a series of specific microbe species had a marked alteration in DAR, accompanied with changes in certain microbial functional pathways.
Collapse
Affiliation(s)
- 一杭 林
- 上海交通大学医学院附属上海儿童医学中心耳鼻咽喉科(上海,200127)Department of Otolaryngology, Shanghai Children's Medical Center of Shanghai Jiao Tong University School of Medcine, Shanghai, 200127, China
| | - 俊阳 李
- 上海交通大学医学院附属上海儿童医学中心耳鼻咽喉科(上海,200127)Department of Otolaryngology, Shanghai Children's Medical Center of Shanghai Jiao Tong University School of Medcine, Shanghai, 200127, China
| | - 幼瑾 李
- 上海交通大学医学院附属上海儿童医学中心耳鼻咽喉科(上海,200127)Department of Otolaryngology, Shanghai Children's Medical Center of Shanghai Jiao Tong University School of Medcine, Shanghai, 200127, China
| |
Collapse
|
31
|
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
|
32
|
Akagawa S, Kaneko K. Gut microbiota and allergic diseases in children. Allergol Int 2022; 71:301-309. [PMID: 35314107 DOI: 10.1016/j.alit.2022.02.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/28/2022] [Accepted: 02/10/2022] [Indexed: 12/11/2022] Open
Abstract
The gut microbiota resides in the human gastrointestinal tract, where it plays an important role in maintaining host health. The human gut microbiota is established by the age of 3 years. Studies have revealed that an imbalance in the gut microbiota, termed dysbiosis, occurs due to factors such as cesarean delivery and antibiotic use before the age of 3 years and that dysbiosis is associated with a higher risk of future onset of allergic diseases. Recent advancements in next-generation sequencing methods have revealed the presence of dysbiosis in patients with allergic diseases, which increases attention on the relationship between dysbiosis and the development of allergic diseases. However, there is no unified perspective on the characteristics on dysbiosis or the mechanistic link between dysbiosis and the onset of allergic diseases. Here, we introduce the latest studies on the gut microbiota in children with allergic diseases and present the hypothesis that dysbiosis characterized by fewer butyric acid-producing bacteria leads to fewer regulatory T cells, resulting in allergic disease. Further studies on correcting dysbiosis for the prevention and treatment of allergic diseases are warranted.
Collapse
Affiliation(s)
- Shohei Akagawa
- Department of Pediatrics, Kansai Medical University, Osaka, Japan
| | - Kazunari Kaneko
- Department of Pediatrics, Kansai Medical University, Osaka, Japan.
| |
Collapse
|
33
|
Lin TL, Wu CY, Fan YH, Chang YL, Ho HJ, Chen YJ. Association between early life laxative exposure and risk of allergic diseases A nationwide matched cohort study. Ann Allergy Asthma Immunol 2022; 128:291-298.e3. [PMID: 34998978 DOI: 10.1016/j.anai.2021.12.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND The early life microbiome can shape human immunity. Recent studies have revealed gut dysbiosis after laxative administration. OBJECTIVE To investigate the impact of infantile laxative exposure on subsequent allergic diseases. METHODS This nationwide matched cohort study was conducted using Taiwan's National Health Insurance Research Database for the period 1997 to 2013. A total of 32,986 patients who had complete information of maternal history and delivery modes were identified. We included 291 children having laxatives for at least 7 days within the first 6 months of life and 1164 reference children not receiving laxatives, matching by sex, propensity score, number of hospital visits, and maternal age at delivery. Demographic characteristics and maternal factors were compared, and cumulative incidences of allergic diseases were calculated. Cox proportional hazard model was used to evaluate associations. RESULTS The 5-year cumulative incidence of allergic diseases in the laxative cohort was significantly higher than that in the reference cohort (49.81% vs 41.68%; P = .01). Early life laxative exposure (adjusted hazard ratio, 1.61; 95% confidence interval, 1.32-1.97) was independently associated with allergic disease development. Other independent risk factors included preterm, male sex, maternal allergic diseases, and prenatal laxative use. Multivariable stratified analyses verified the association between early life laxative exposure and subsequent allergic disease development in all subgroups of children, including those born to mothers without allergic diseases or prenatal laxative use. CONCLUSION Early life laxative exposure is associated with allergic disease development.
Collapse
Affiliation(s)
- Teng-Li Lin
- Department of Dermatology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
| | - Chun-Ying Wu
- Institute of Biomedical Informatics and Institute of Public Health, National Yang Ming Chiao Tung University, Taipei, Taiwan; Faculty of Medicine and Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Division of Translational Research, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Public Health, China Medical University, Taichung, Taiwan; National Institute of Cancer Research and Institute of Population Health Science, National Health Research Institutes, Miaoli, Taiwan
| | - Yi-Hsuan Fan
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yi-Ling Chang
- Department of Dermatology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Hsiu J Ho
- Institute of Biomedical Informatics and Institute of Public Health, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Ju Chen
- Faculty of Medicine and Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Dermatology, Taichung Veterans General Hospital, Taichung, Taiwan.
| |
Collapse
|
34
|
Lee MJ, Park YM, Kim B, Tae IH, Kim NE, Pranata M, Kim T, Won S, Kang NJ, Lee YK, Lee DW, Nam MH, Hong SJ, Kim BS. Disordered development of gut microbiome interferes with the establishment of the gut ecosystem during early childhood with atopic dermatitis. Gut Microbes 2022; 14:2068366. [PMID: 35485368 PMCID: PMC9067516 DOI: 10.1080/19490976.2022.2068366] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 04/07/2022] [Accepted: 04/11/2022] [Indexed: 02/04/2023] Open
Abstract
The gut microbiome influences the development of allergic diseases during early childhood. However, there is a lack of comprehensive understanding of microbiome-host crosstalk. Here, we analyzed the influence of gut microbiome dynamics in early childhood on atopic dermatitis (AD) and the potential interactions between host and microbiome that control this homeostasis. We analyzed the gut microbiome in 346 fecal samples (6-36 months; 112 non-AD, 110 mild AD, and 124 moderate to severe AD) from the Longitudinal Cohort for Childhood Origin of Asthma and Allergic Disease birth cohort. The microbiome-host interactions were analyzed in animal and in vitro cell assays. Although the gut microbiome maturated with age in both AD and non-AD groups, its development was disordered in the AD group. Disordered colonization of short-chain fatty acids (SCFA) producers along with age led to abnormal SCFA production and increased IgE levels. A butyrate deficiency and downregulation of GPR109A and PPAR-γ genes were detected in AD-induced mice. Insufficient butyrate decreases the oxygen consumption rate of host cells, which can release oxygen to the gut and perturb the gut microbiome. The disordered gut microbiome development could aggravate balanced microbiome-host interactions, including immune responses during early childhood with AD.
Collapse
Affiliation(s)
- Min-Jung Lee
- Department of Life Science, Multidisciplinary Genome Institute, Hallym University, Chuncheon, Republic of Korea
| | - Yoon Mee Park
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Byunghyun Kim
- Seoul Center, Korea Basic Science Institute, Seoul, Republic of Korea
| | - in Hwan Tae
- Seoul Center, Korea Basic Science Institute, Seoul, Republic of Korea
| | - Nam-Eun Kim
- Department of Public Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Marina Pranata
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bioscience, Soonchunhyang University, Cheonan, Republic of Korea
| | - Taewon Kim
- School of Food Science and Biotechnology, Kyungpook National University, Daegu, Republic of Korea
| | - Sungho Won
- Department of Public Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Nam Joo Kang
- School of Food Science and Biotechnology, Kyungpook National University, Daegu, Republic of Korea
- Department of Integrative Biology, Kyungpook National University, Daegu, Republic of Korea
| | - Yun Kyung Lee
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bioscience, Soonchunhyang University, Cheonan, Republic of Korea
| | - Dong-Woo Lee
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Myung Hee Nam
- Seoul Center, Korea Basic Science Institute, Seoul, Republic of Korea
| | - Soo-Jong Hong
- Department of Pediatrics, Childhood Asthma Atopy Center, Humidifier Disinfectant Health Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Bong-Soo Kim
- Department of Life Science, Multidisciplinary Genome Institute, Hallym University, Chuncheon, Republic of Korea
- The Korean Institute of Nutrition, Hallym University, Chuncheon, Republic of Korea
| |
Collapse
|
35
|
Akagawa S, Akagawa Y, Yamanouchi S, Teramoto Y, Yasuda M, Fujishiro S, Kino J, Hirabayashi M, Mine K, Kimata T, Hashiyada M, Akane A, Tsuji S, Kaneko K. Association of Neonatal Jaundice with Gut Dysbiosis Characterized by Decreased Bifidobacteriales. Metabolites 2021; 11:metabo11120887. [PMID: 34940645 PMCID: PMC8705620 DOI: 10.3390/metabo11120887] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 12/15/2021] [Indexed: 11/16/2022] Open
Abstract
Neonatal jaundice, caused by excess serum bilirubin levels, is a common condition in neonates. Imbalance in the gut microbiota is believed to play a role in the development of neonatal jaundice. Thus, we aimed to reveal the gut microbiota characteristics in neonates with jaundice. 16S rRNA gene sequencing was performed on stool samples collected on day 4 from 26 neonates with jaundice (serum total bilirubin > 15.0 mg/dL) and 17 neonates without jaundice (total serum bilirubin < 10.0 mg/dL). All neonates were born full term, with normal weight, by vaginal delivery, and were breastfed. Neonates who were administered antibiotics, had serum direct bilirubin levels above 1 mg/dL, or had conditions possibly leading to hemolytic anemia were excluded. The median serum bilirubin was 16.0 mg/dL (interquartile range: 15.5-16.8) and 7.4 mg/dL (interquartile range: 6.8-8.3) for the jaundice and non-jaundice groups, respectively. There was no difference in the alpha diversity indices. Meanwhile, in the jaundice group, linear discriminant analysis effect size revealed that Bifidobacteriales were decreased at the order level, while Enterococcaceae were increased and Bifidobacteriaceae were decreased at the family level. Bifidobacteriaceae may act preventatively because of their suppressive effect on beta-glucuronidase, leading to accelerated deconjugation of conjugated bilirubin in the intestine. In summary, neonates with jaundice had dysbiosis characterized by a decreased abundance of Bifidobacteriales.
Collapse
Affiliation(s)
- Shohei Akagawa
- Department of Pediatrics, Kansai Medical University, Hirakata-shi, Osaka 573-1010, Japan; (S.A.); (Y.A.); (S.Y.); (Y.T.); (M.Y.); (S.F.); (J.K.); (M.H.); (K.M.); (T.K.); (S.T.)
| | - Yuko Akagawa
- Department of Pediatrics, Kansai Medical University, Hirakata-shi, Osaka 573-1010, Japan; (S.A.); (Y.A.); (S.Y.); (Y.T.); (M.Y.); (S.F.); (J.K.); (M.H.); (K.M.); (T.K.); (S.T.)
| | - Sohsaku Yamanouchi
- Department of Pediatrics, Kansai Medical University, Hirakata-shi, Osaka 573-1010, Japan; (S.A.); (Y.A.); (S.Y.); (Y.T.); (M.Y.); (S.F.); (J.K.); (M.H.); (K.M.); (T.K.); (S.T.)
| | - Yoshiki Teramoto
- Department of Pediatrics, Kansai Medical University, Hirakata-shi, Osaka 573-1010, Japan; (S.A.); (Y.A.); (S.Y.); (Y.T.); (M.Y.); (S.F.); (J.K.); (M.H.); (K.M.); (T.K.); (S.T.)
| | - Masahiro Yasuda
- Department of Pediatrics, Kansai Medical University, Hirakata-shi, Osaka 573-1010, Japan; (S.A.); (Y.A.); (S.Y.); (Y.T.); (M.Y.); (S.F.); (J.K.); (M.H.); (K.M.); (T.K.); (S.T.)
| | - Sadayuki Fujishiro
- Department of Pediatrics, Kansai Medical University, Hirakata-shi, Osaka 573-1010, Japan; (S.A.); (Y.A.); (S.Y.); (Y.T.); (M.Y.); (S.F.); (J.K.); (M.H.); (K.M.); (T.K.); (S.T.)
| | - Jiro Kino
- Department of Pediatrics, Kansai Medical University, Hirakata-shi, Osaka 573-1010, Japan; (S.A.); (Y.A.); (S.Y.); (Y.T.); (M.Y.); (S.F.); (J.K.); (M.H.); (K.M.); (T.K.); (S.T.)
| | - Masato Hirabayashi
- Department of Pediatrics, Kansai Medical University, Hirakata-shi, Osaka 573-1010, Japan; (S.A.); (Y.A.); (S.Y.); (Y.T.); (M.Y.); (S.F.); (J.K.); (M.H.); (K.M.); (T.K.); (S.T.)
| | - Kenji Mine
- Department of Pediatrics, Kansai Medical University, Hirakata-shi, Osaka 573-1010, Japan; (S.A.); (Y.A.); (S.Y.); (Y.T.); (M.Y.); (S.F.); (J.K.); (M.H.); (K.M.); (T.K.); (S.T.)
| | - Takahisa Kimata
- Department of Pediatrics, Kansai Medical University, Hirakata-shi, Osaka 573-1010, Japan; (S.A.); (Y.A.); (S.Y.); (Y.T.); (M.Y.); (S.F.); (J.K.); (M.H.); (K.M.); (T.K.); (S.T.)
| | - Masaki Hashiyada
- Department of Legal Medicine, Kansai Medical University, Hirakata-shi, Osaka 573-1010, Japan; (M.H.); (A.A.)
| | - Atsushi Akane
- Department of Legal Medicine, Kansai Medical University, Hirakata-shi, Osaka 573-1010, Japan; (M.H.); (A.A.)
| | - Shoji Tsuji
- Department of Pediatrics, Kansai Medical University, Hirakata-shi, Osaka 573-1010, Japan; (S.A.); (Y.A.); (S.Y.); (Y.T.); (M.Y.); (S.F.); (J.K.); (M.H.); (K.M.); (T.K.); (S.T.)
| | - Kazunari Kaneko
- Department of Pediatrics, Kansai Medical University, Hirakata-shi, Osaka 573-1010, Japan; (S.A.); (Y.A.); (S.Y.); (Y.T.); (M.Y.); (S.F.); (J.K.); (M.H.); (K.M.); (T.K.); (S.T.)
- Correspondence: ; Tel.: +81-72-804-0101
| |
Collapse
|
36
|
Bodden C, Pang TY, Feng Y, Mridha F, Kong G, Li S, Watt MJ, Reichelt AC, Hannan AJ. Intergenerational effects of a paternal Western diet during adolescence on offspring gut microbiota, stress reactivity, and social behavior. FASEB J 2021; 36:e21981. [PMID: 34907601 DOI: 10.1096/fj.202100920rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 09/16/2021] [Accepted: 09/22/2021] [Indexed: 12/28/2022]
Abstract
The global consumption of highly processed, calorie-dense foods has contributed to an epidemic of overweight and obesity, along with negative consequences for metabolic dysfunction and disease susceptibility. As it becomes apparent that overweight and obesity have ripple effects through generations, understanding of the processes involved is required, in both maternal and paternal epigenetic inheritance. We focused on the patrilineal effects of a Western-style high-fat (21%) and high-sugar (34%) diet (WD) compared to control diet (CD) during adolescence and investigated F0 and F1 mice for physiological and behavioral changes. F0 males (fathers) showed increased body weight, impaired glycemic control, and decreased attractiveness to females. Paternal WD caused significant phenotypic changes in F1 offspring, including higher body weights of pups, increased Actinobacteria abundance in the gut microbiota (ascertained using 16S microbiome profiling), a food preference for WD pellets, increased male dominance and attractiveness to females, as well as decreased behavioral despair. These results collectively demonstrate the long-term intergenerational effects of a Western-style diet during paternal adolescence. The behavioral and physiological alterations in F1 offspring provide evidence of adaptive paternal programming via epigenetic inheritance. These findings have important implications for understanding paternally mediated intergenerational inheritance, and its relevance to offspring health and disease susceptibility.
Collapse
Affiliation(s)
- Carina Bodden
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Terence Y Pang
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Yingshi Feng
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Faria Mridha
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Geraldine Kong
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Shanshan Li
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Matthew J Watt
- Department of Anatomy and Physiology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Amy C Reichelt
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia.,Department of Medical Sciences, Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Anthony J Hannan
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia.,Department of Anatomy and Physiology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
37
|
Saturio S, Nogacka AM, Alvarado-Jasso GM, Salazar N, de los Reyes-Gavilán CG, Gueimonde M, Arboleya S. Role of Bifidobacteria on Infant Health. Microorganisms 2021; 9:2415. [PMID: 34946017 PMCID: PMC8708449 DOI: 10.3390/microorganisms9122415] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/19/2021] [Accepted: 11/21/2021] [Indexed: 12/19/2022] Open
Abstract
Bifidobacteria are among the predominant microorganisms during infancy, being a dominant microbial group in the healthy breastfed infant and playing a crucial role in newborns and infant development. Not only the levels of the Bifidobacterium genus but also the profile and quantity of the different bifidobacterial species have been demonstrated to be of relevance to infant health. Although no definitive proof is available on the causal association, reduced levels of bifidobacteria are perhaps the most frequently observed alteration of the intestinal microbiota in infant diseases. Moreover, Bifidobacterium strains have been extensively studied by their probiotic attributes. This review compiles the available information about bifidobacterial composition and function since the beginning of life, describing different perinatal factors affecting them, and their implications on different health alterations in infancy. In addition, this review gathers exhaustive information about pre-clinical and clinical studies with Bifidobacterium strains as probiotics in neonates.
Collapse
Affiliation(s)
- Silvia Saturio
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.M.N.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.-G.)
- Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
| | - Alicja M. Nogacka
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.M.N.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.-G.)
- Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
| | - Guadalupe M. Alvarado-Jasso
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.M.N.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.-G.)
| | - Nuria Salazar
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.M.N.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.-G.)
- Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
| | - Clara G. de los Reyes-Gavilán
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.M.N.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.-G.)
- Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.M.N.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.-G.)
- Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
| | - Silvia Arboleya
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.M.N.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.-G.)
- Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
| |
Collapse
|
38
|
Feng WW, Liu J, Cheng H, Peng C. Integration of Gut Microbiota and Metabolomics for Chinese Medicines Research: Opportunities and Challenges. Chin J Integr Med 2021; 28:1032-1039. [PMID: 34755290 DOI: 10.1007/s11655-021-3305-x] [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] [Accepted: 01/28/2021] [Indexed: 12/15/2022]
Abstract
Chinese medicines (CM) are gaining more attentions from all over the world. However, there are a large body of questions to be answered because of the chemical complexity of CM and intricate molecular reactions within human body. In recent years, gut microbiota and metabolomics have emerged as two cynosures in deciphering the mechanisms of how our body is functioning. Since gut microbiota and host is a closely interrelated system, paying attention only to gut microbiota or metabolites may omit the interplays among CM, gut microbiota, and hosts. To systemically study these interplays, a network understanding of CM components, gut microbiota, metabolites of gut microbiota, metabolites in human body is necessary. Although there are some obstacles impeding the application of this integrative approach, the potential areas for implementation of the integrative approach is vast. These areas include, but not limited to, elucidating the mechanisms of CM at system level, screening bioactive compounds in CM, and guiding quality control of CM.
Collapse
Affiliation(s)
- Wu-Wen Feng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Juan Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Hao Cheng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Cheng Peng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
39
|
Xiang K, Wang P, Xu Z, Hu YQ, He YS, Chen Y, Feng YT, Yin KJ, Huang JX, Wang J, Wu ZD, Yang XK, Wang DG, Ye DQ, Pan HF. Causal Effects of Gut Microbiome on Systemic Lupus Erythematosus: A Two-Sample Mendelian Randomization Study. Front Immunol 2021; 12:667097. [PMID: 34557183 PMCID: PMC8453215 DOI: 10.3389/fimmu.2021.667097] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 08/16/2021] [Indexed: 12/19/2022] Open
Abstract
The observational association between gut microbiome and systemic lupus erythematosus (SLE) has been well documented. However, whether the association is causal remains unclear. The present study used publicly available genome-wide association study (GWAS) summary data to perform two-sample Mendelian randomization (MR), aiming to examine the causal links between gut microbiome and SLE. Two sets of MR analyses were conducted. A group of single nucleotide polymorphisms (SNPs) that less than the genome-wide statistical significance threshold (5 × 10-8) served as instrumental variables. To obtain a comprehensive conclusion, the other group where SNPs were smaller than the locus-wide significance level (1 × 10-5) were selected as instrumental variables. Based on the locus-wide significance level, the results indicated that there were causal effects of gut microbiome components on SLE risk. The inverse variance weighted (IVW) method suggested that Bacilli and Lactobacillales were positively correlated with the risk of SLE and Bacillales, Coprobacter and Lachnospira were negatively correlated with SLE risk. The results of weighted median method supported that Bacilli, Lactobacillales, and Eggerthella were risk factors for SLE and Bacillales and Coprobacter served as protective factors for SLE. The estimates of MR Egger suggested that genetically predicted Ruminiclostridium6 was negatively associated with SLE. Based on the genome-wide statistical significance threshold, the results showed that Actinobacteria might reduce the SLE risk. However, Mendelian randomization pleiotropy residual sum and outlier (MR-PRESSO) detected significant horizontal pleiotropy between the instrumental variables of Ruminiclostridium6 and outcome. This study support that there are beneficial or detrimental causal effects of gut microbiome components on SLE risk.
Collapse
Affiliation(s)
- Kun Xiang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Peng Wang
- Center for Genetic Epidemiology and Genomics, School of Public Health, Soochow University Medical College, Suzhou, China
| | - Zhiwei Xu
- School of Public Health, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Yu-Qian Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Yi-Sheng He
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Yue Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Ya-Ting Feng
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Kang-Jia Yin
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Ji-Xiang Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Jie Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Zheng-Dong Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Xiao-Ke Yang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - De-Guang Wang
- Department of Nephrology, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Dong-Qing Ye
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| |
Collapse
|
40
|
Baur R, Gandhi J, Marshall NB, Lukomska E, Weatherly LM, Shane HL, Hu G, Anderson SE. Dermal exposure to the immunomodulatory antimicrobial chemical triclosan alters the skin barrier integrity and microbiome in mice. Toxicol Sci 2021; 184:223-235. [PMID: 34515797 DOI: 10.1093/toxsci/kfab111] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Triclosan is an antimicrobial chemical used in healthcare settings that can be absorbed through the skin. Exposure to triclosan has been positively associated with food and aeroallergy and asthma exacerbation in humans and, although not directly sensitizing, has been demonstrated to augment the allergic response in a mouse model of asthma. The skin barrier and microbiome are thought to play important roles in regulating inflammation and allergy and disruptions may contribute to development of allergic disease. To investigate potential connections of the skin barrier and microbiome with immune responses to triclosan, SKH1 mice were exposed dermally to triclosan (0.5-2%) or vehicle for up to 7 consecutive days. Exposure to 2% triclosan for 5-7 days on the skin was shown to increase trans-epidermal water loss levels. Seven days of dermal exposure to triclosan decreased filaggrin 2 and keratin 10 expression, but increased filaggrin and keratin 14 protein along with the danger signal S100a8 and interleukin-4. Dermal exposure to triclosan for 7 days also altered the alpha and beta diversity of the skin and gut microbiome. Specifically, dermal triclosan exposure increased the relative abundance of the Firmicutes family, Lachnospiraceae on the skin but decreased the abundance of Firmicutes family, Ruminococcaceae in the gut. Collectively, these results demonstrate that repeated dermal exposure to the antimicrobial chemical triclosan alters the skin barrier integrity and microbiome in mice, suggesting that these changes may contribute to the increase in allergic immune responses following dermal exposure to triclosan.
Collapse
Affiliation(s)
- Rachel Baur
- Allergy and Clinical Immunology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV.,Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University, Morgantown, WV
| | - Jasleen Gandhi
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University, Morgantown, WV
| | - Nikki B Marshall
- Allergy and Clinical Immunology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV
| | - Ewa Lukomska
- Allergy and Clinical Immunology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV
| | - Lisa M Weatherly
- Allergy and Clinical Immunology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV
| | - Hillary L Shane
- Allergy and Clinical Immunology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV
| | - Gangqing Hu
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University, Morgantown, WV.,WVU Cancer Institute, West Virginia University, Morgantown, WV, 26506, USA
| | - Stacey E Anderson
- Allergy and Clinical Immunology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV
| |
Collapse
|
41
|
Schei K, Simpson MR, Øien T, Salamati S, Rudi K, Ødegård RA. Allergy-related diseases and early gut fungal and bacterial microbiota abundances in children. Clin Transl Allergy 2021; 11:e12041. [PMID: 34194728 PMCID: PMC8238386 DOI: 10.1002/clt2.12041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 05/07/2021] [Accepted: 06/16/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND The early gut microbiota has been proposed as an important link between environmental exposures and development of allergy-related diseases. Beyond the widely investigated associations between the gut bacterial microbiota, we investigated the involvement of early gut mycobiota and gut permeability in the pathogenesis of asthma, allergic rhinoconjunctivitis (AR) and eczema. METHODS In the Probiotics in the Prevention of Allergy among Children in Trondheim trial with maternal probiotic supplementation, we collected faecal samples at four timepoints between 0 and 2 years from a cohort of 278 children. Clinical information on allergy-related diseases was collected in a paediatric examination at 2 years and questionnaires at 6 weeks and 1, 2 and 6 years. By quantitative PCR and 16S/ITS1 MiSeq rRNA gene sequencing, we analysed the gut bacterial and fungal microbiota abundance and bacterial diversity and explored associations with allergy-related diseases. We also measured gut permeability markers (lipopolysaccharide-binding protein [LBP] and fatty acid-binding protein 2 [FABP2]). RESULTS Children with higher fungal abundance at 2 years were more likely to develop asthma and AR by 6 years, odds ratios 1.70 (95% CI: 1.06-2.75) and 1.41 (1.03-1.93), respectively. We explored causal connections, and children with eczema at 1-2 years appeared to have more mature bacterial microbiota, as well as being depleted of Enterococcus genus. Although LBP and FABP2 did not correlate with eczema, increased bacterial abundance was associated with increased serum FABP2. CONCLUSIONS We observed positive associations between gut fungal abundance and allergy-related disease, but increased gut permeability does not appear to be involved in the underlying mechanisms for this association. Our findings should be confirmed in future microbiota studies.
Collapse
Affiliation(s)
- Kasper Schei
- Department of Clinical and Molecular MedicineFaculty of Medicine and Health SciencesNTNU–Norwegian University of Science and TechnologyTrondheimNorway
| | - Melanie Rae Simpson
- Department of Public Health and NursingFaculty of Medicine and Health SciencesNTNU–Norwegian University of Science and TechnologyTrondheimNorway
- Clinic of Laboratory MedicineSt. Olav's HospitalTrondheimNorway
| | - Torbjørn Øien
- Department of Public Health and NursingFaculty of Medicine and Health SciencesNTNU–Norwegian University of Science and TechnologyTrondheimNorway
| | - Saideh Salamati
- Regional Centre of Obesity Research and Innovation (ObeCe)St. Olav's HospitalTrondheimNorway
| | - Knut Rudi
- Faculty of Chemistry, Biotechnology and Food ScienceNorwegian University of Life SciencesÅsNorway
| | - Rønnaug Astri Ødegård
- Department of Clinical and Molecular MedicineFaculty of Medicine and Health SciencesNTNU–Norwegian University of Science and TechnologyTrondheimNorway
- Regional Centre of Obesity Research and Innovation (ObeCe)St. Olav's HospitalTrondheimNorway
| |
Collapse
|
42
|
Li X, Peng Y, Li Z, Christensen B, Heckmann AB, Lagerqvist C, Stenlund H, Lönnerdal B, Hernell O, West CE. Serum cytokine patterns are modulated in infants fed formula with probiotics or milk fat globule membranes: A randomized controlled trial. PLoS One 2021; 16:e0251293. [PMID: 33983993 PMCID: PMC8118299 DOI: 10.1371/journal.pone.0251293] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 03/12/2021] [Indexed: 01/27/2023] Open
Abstract
Background Proteins and lipids of milk fat globule membrane (MFGM) and probiotics are immunomodulatory. We hypothesized that Lactobacillus paracasei ssp. paracasei strain F19 (F19) would augment vaccine antibody and T helper 1 type immune responses whereas MFGM would produce an immune response closer to that of breastfed (BF) infants. Objective To compare the effects of supplementing formula with F19 or bovine MFGM on serum cytokine and vaccine responses of formula-fed (FF) and BF infants. Design FF infants were randomized to formula with F19 (n = 195) or MFGM (n = 192), or standard formula (SF) (n = 194) from age 21±7 days until 4 months. A BF group served as reference (n = 208). We analyzed seven cytokines (n = 398) in serum at age 4 months using magnetic bead-based multiplex technology. Using ELISA, we analyzed anti-diphtheria IgG (n = 258) and anti-poliovirus IgG (n = 309) concentrations in serum before and after the second and third immunization, respectively. Results Compared with SF, the F19 group had greater IL-2 and lower IFN-γ concentrations (p<0.05, average effect size 0.14 and 0.39). Compared with BF, the F19 group had greater IL-2, IL-4 and IL-17A concentrations (p<0.05, average effect size 0.42, 0.34 and 0.26, respectively). The MFGM group had lower IL-2 and IL-17A concentrations compared with SF (p<0.05, average effect size 0.34 and 0.31). Cytokine concentrations were comparable among the MFGM and BF groups. Vaccine responses were comparable among the formula groups. Conclusions Contrary to previous studies F19 increased IL-2 and lowered IFN-γ production, suggesting that the response to probiotics differs across populations. The cytokine profile of the MFGM group approached that of BF infants, and may be associated with the previous finding that infectious outcomes for the MFGM group in this cohort were closer to those of BF infants, as opposed to the SF group. These immunomodulatory effects support future clinical evaluation of infant formula with F19 or MFGM.
Collapse
Affiliation(s)
- Xiaonan Li
- Department of Child Health Care, Childrens Hospital of Nanjing Medical University, Nanjing, China
| | - Yongmei Peng
- Department of Children Health Care, Childrens Hospital of Fudan University, Shanghai, China
| | - Zailing Li
- Department of Pediatrics, Beijing University Third Hospital, Bejing, China
| | - Britt Christensen
- Arla Innovation Center, Arla Foods amba, Skejby, Denmark
- * E-mail: (CEW); (BC)
| | | | - Carina Lagerqvist
- Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, Sweden
| | - Hans Stenlund
- Departments of Public Health and Clinical Medicine, Epidemiology and Global Health, Umeå University, Umeå, Sweden
| | - Bo Lönnerdal
- Department of Nutrition, University of California, Davis, California, United States of America
| | - Olle Hernell
- Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, Sweden
| | - Christina E. West
- Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, Sweden
- * E-mail: (CEW); (BC)
| |
Collapse
|
43
|
Wang S, Egan M, Ryan CA, Boyaval P, Dempsey EM, Ross RP, Stanton C. A good start in life is important-perinatal factors dictate early microbiota development and longer term maturation. FEMS Microbiol Rev 2021; 44:763-781. [PMID: 32821932 PMCID: PMC7685781 DOI: 10.1093/femsre/fuaa030] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 08/18/2020] [Indexed: 12/20/2022] Open
Abstract
Maternal health status is vital for the development of the offspring of humans, including physiological health and psychological functions. The complex and diverse microbial ecosystem residing within humans contributes critically to these intergenerational impacts. Perinatal factors, including maternal nutrition, antibiotic use and maternal stress, alter the maternal gut microbiota during pregnancy, which can be transmitted to the offspring. In addition, gestational age at birth and mode of delivery are indicated frequently to modulate the acquisition and development of gut microbiota in early life. The early-life gut microbiota engages in a range of host biological processes, particularly immunity, cognitive neurodevelopment and metabolism. The perturbed early-life gut microbiota increases the risk for disease in early and later life, highlighting the importance of understanding relationships of perinatal factors with early-life microbial composition and functions. In this review, we present an overview of the crucial perinatal factors and summarise updated knowledge of early-life microbiota, as well as how the perinatal factors shape gut microbiota in short and long terms. We further discuss the clinical consequences of perturbations of early-life gut microbiota and potential therapeutic interventions with probiotics/live biotherapeutics.
Collapse
Affiliation(s)
- Shaopu Wang
- APC Microbiome Ireland, Cork, Ireland, P12 YT20.,Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland, P61 C996
| | - Muireann Egan
- APC Microbiome Ireland, Cork, Ireland, P12 YT20.,Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland, P61 C996
| | - C Anthony Ryan
- APC Microbiome Ireland, Cork, Ireland, P12 YT20.,Department of Paediatrics & Child Health, University College Cork, Cork, Ireland, T12 YN60
| | - Patrick Boyaval
- DuPont Nutrition & Biosciences, Danisco France SAS - DuPont, 22, rue Brunel, F- 75017 Paris, France
| | - Eugene M Dempsey
- APC Microbiome Ireland, Cork, Ireland, P12 YT20.,Department of Paediatrics & Child Health, University College Cork, Cork, Ireland, T12 YN60
| | - R Paul Ross
- APC Microbiome Ireland, Cork, Ireland, P12 YT20
| | - Catherine Stanton
- APC Microbiome Ireland, Cork, Ireland, P12 YT20.,Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland, P61 C996
| |
Collapse
|
44
|
Chua RXY, Tay MJY, Ooi DSQ, Siah KTH, Tham EH, Shek LPC, Meaney MJ, Broekman BFP, Loo EXL. Understanding the Link Between Allergy and Neurodevelopmental Disorders: A Current Review of Factors and Mechanisms. Front Neurol 2021; 11:603571. [PMID: 33658968 PMCID: PMC7917177 DOI: 10.3389/fneur.2020.603571] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 12/30/2020] [Indexed: 12/17/2022] Open
Abstract
Both allergic diseases and neurodevelopmental disorders are non-communicable diseases (NCDs) that not only impact on the quality of life and but also result in substantial economic burden. Immune dysregulation and inflammation are typical hallmarks in both allergic and neurodevelopmental disorders, suggesting converging pathophysiology. Epidemiological studies provided convincing evidence for the link between allergy and neurodevelopmental diseases such as attention-deficit hyperactivity disorder (ADHD) and autism spectrum disorder (ASD). Possible factors influencing the development of these disorders include maternal depression and anxiety, gestational diabetes mellitus, maternal allergic status, diet, exposure to environmental pollutants, microbiome dysbiosis, and sleep disturbances that occur early in life. Moreover, apart from inflammation, epigenetics, gene expression, and mitochondrial dysfunction have emerged as possible underlying mechanisms in the pathogenesis of these conditions. The exploration and understanding of these shared factors and possible mechanisms may enable us to elucidate the link in the comorbidity.
Collapse
Affiliation(s)
- Regena Xin Yi Chua
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Michelle Jia Yu Tay
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Delicia Shu Qin Ooi
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, National University Health System, Singapore, Singapore
| | - Kewin Tien Ho Siah
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Division of Gastroenterology & Hepatology, University Medicine Cluster, National University Hospital, Singapore, Singapore
| | - Elizabeth Huiwen Tham
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, National University Health System, Singapore, Singapore.,Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Lynette Pei-Chi Shek
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, National University Health System, Singapore, Singapore.,Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Michael J Meaney
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore.,Ludmer Centre for Neuroinformatics and Mental Health and Department of Psychiatry, McGill University, Montréal, QC, Canada
| | - Birit F P Broekman
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore.,Department of Psychiatry, Onze Lieve Vrouwe Gasthuis and Amsterdam University Medical Centre, VU University Medical Center, Amsterdam, Netherlands
| | - Evelyn Xiu Ling Loo
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| |
Collapse
|
45
|
Egan M, Dempsey E, Ryan CA, Ross RP, Stanton C. The Sporobiota of the Human Gut. Gut Microbes 2021; 13:1-17. [PMID: 33406976 PMCID: PMC7801112 DOI: 10.1080/19490976.2020.1863134] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 12/01/2020] [Accepted: 12/04/2020] [Indexed: 02/04/2023] Open
Abstract
The human gut microbiome is a diverse and complex ecosystem that plays a critical role in health and disease. The composition of the gut microbiome has been well studied across all stages of life. In recent years, studies have investigated the production of endospores by specific members of the gut microbiome. An endospore is a tough, dormant structure formed by members of the Firmicutes phylum, which allows for greater resistance to otherwise inhospitable conditions. This innate resistance has consequences for human health and disease, as well as in biotechnology. In particular, the formation of endospores is strongly linked to antibiotic resistance and the spread of antibiotic resistance genes, also known as the resistome. The term sporobiota has been used to define the spore-forming cohort of a microbial community. In this review, we present an overview of the current knowledge of the sporobiota in the human gut. We discuss the development of the sporobiota in the infant gut and the perinatal factors that may have an effect on vertical transmission from mother to infant. Finally, we examine the sporobiota of critically important food sources for the developing infant, breast milk and powdered infant formula.
Collapse
Affiliation(s)
- Muireann Egan
- Food Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Eugene Dempsey
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Neonatology, Cork University Maternity Hospital, Cork, Ireland
| | - C. Anthony Ryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Neonatology, Cork University Maternity Hospital, Cork, Ireland
| | - R. Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Catherine Stanton
- Food Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
46
|
Li L, Fang Z, Liu Z, Zhao J, Zhang H, Wang S, He J, Lu W, Chen W. Lactobacillus reuteri CCFM1072 and CCFM1040 with the role of Treg cells regulation alleviate airway inflammation through modulating gut microbiota in allergic asthma mice. J Funct Foods 2021. [DOI: 10.1016/j.jff.2020.104286] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
47
|
|
48
|
Alkotob SS, Cannedy C, Harter K, Movassagh H, Paudel B, Prunicki M, Sampath V, Schikowski T, Smith E, Zhao Q, Traidl‐Hoffmann C, Nadeau KC. Advances and novel developments in environmental influences on the development of atopic diseases. Allergy 2020; 75:3077-3086. [PMID: 33037680 DOI: 10.1111/all.14624] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 10/05/2020] [Accepted: 10/05/2020] [Indexed: 12/12/2022]
Abstract
Although genetic factors play a role in the etiology of atopic disease, the rapid increases in the prevalence of these diseases over the last few decades suggest that environmental, rather than genetic factors are the driving force behind the increasing prevalence. In modern societies, there is increased time spent indoors, use of antibiotics, and consumption of processed foods and decreased contact with farm animals and pets, which limit exposure to environmental allergens, infectious parasitic worms, and microbes. The lack of exposure to these factors is thought to prevent proper education and training of the immune system. Increased industrialization and urbanization have brought about increases in organic and inorganic pollutants. In addition, Caesarian birth, birth order, increased use of soaps and detergents, tobacco smoke exposure and psychosomatic factors are other factors that have been associated with increased rate of allergic diseases. Here, we review current knowledge on the environmental factors that have been shown to affect the development of allergic diseases and the recent developments in the field.
Collapse
Affiliation(s)
- Shifaa Suhayl Alkotob
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University and Division of Pulmonary and Critical Care Medicine Department of Medicine Stanford University Stanford CA USA
| | - Cade Cannedy
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University and Division of Pulmonary and Critical Care Medicine Department of Medicine Stanford University Stanford CA USA
| | - Katharina Harter
- Chair and Institute of Environmental Medicine UNIKA‐TTechnical University of Munich and Helmholtz Zentrum München Augsburg Germany
| | - Hesam Movassagh
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University and Division of Pulmonary and Critical Care Medicine Department of Medicine Stanford University Stanford CA USA
| | - Bibek Paudel
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University and Division of Pulmonary and Critical Care Medicine Department of Medicine Stanford University Stanford CA USA
| | - Mary Prunicki
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University and Division of Pulmonary and Critical Care Medicine Department of Medicine Stanford University Stanford CA USA
| | - Vanitha Sampath
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University and Division of Pulmonary and Critical Care Medicine Department of Medicine Stanford University Stanford CA USA
| | - Tamara Schikowski
- IUF‐Leibniz Institute for Environmental Medicine Duesseldorf Germany
| | - Eric Smith
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University and Division of Pulmonary and Critical Care Medicine Department of Medicine Stanford University Stanford CA USA
| | - Qi Zhao
- IUF‐Leibniz Institute for Environmental Medicine Duesseldorf Germany
| | - Claudia Traidl‐Hoffmann
- Chair and Institute of Environmental Medicine UNIKA‐TTechnical University of Munich and Helmholtz Zentrum München Augsburg Germany
- CK‐CARE Christine Kühne Center for Allergy Research and Education Davos Switzerland
| | - Kari C. Nadeau
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University and Division of Pulmonary and Critical Care Medicine Department of Medicine Stanford University Stanford CA USA
| |
Collapse
|
49
|
Han X, Krempski JW, Nadeau K. Advances and novel developments in mechanisms of allergic inflammation. Allergy 2020; 75:3100-3111. [PMID: 33068299 DOI: 10.1111/all.14632] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/13/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023]
Abstract
In the past decade, research in the molecular and cellular underpinnings of basic and clinical immunology has significantly advanced our understanding of allergic disorders, allowing scientists and clinicians to diagnose and treat disorders such as asthma, allergic and nonallergic rhinitis, and food allergy. In this review, we discuss several significant recent developments in basic and clinical research as well as important future research directions in allergic inflammation. Certain key regulatory cytokines, genes and molecules have recently been shown to play key roles in allergic disorders. For example, interleukin-33 (IL-33) plays an important role in refractory disorders such as asthma, allergic rhinitis and food allergy, mainly by inducing T helper (Th) 2 immune responses and clinical trials with IL-33 inhibitors are underway in food allergy. We discuss interleukin 4 receptor pathways, which recently have been shown to play a critical role among the allergic inflammatory pathways that drive allergic disorders and pathogenesis. Further, the cytokine thymic stromal lymphopoietin (TSLP) has recently been shown as a factor in maintaining immune homeostasis and regulating type 2 inflammatory responses at mucosal barriers in allergic inflammation and targeting TSLP-mediated signalling is considered an attractive therapeutic strategy. In addition, new findings establish an important T cell-intrinsic role of mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1) proteolytic activity in the suppression of autoimmune responses. We have seen how mutations in the filaggrin gene are a significant risk factor for allergic diseases such as atopic dermatitis, asthma, allergic rhinitis, food allergy, contact allergy, and hand eczema. We are only beginning to understand the mechanisms by which the human microbiota may be regulating the immune system, and how sudden changes in the composition of the microbiota may have profound effects, linked with an increased risk of developing chronic inflammatory disorders, including allergies. New research has shown the important but complex role monocytes play in disorders such as food allergies. Finally, we discuss some of the new directions of research in this area, particularly the important use of biologicals in oral immunotherapy, advances in gene therapy, multifood therapy, novel diagnostics in diagnosing allergic disorders and the central role that omics play in creating molecular signatures and biomarkers of allergic disorders such as food allergy. Such exciting new developments and advances have significantly moved forth our ability to understand the mechanisms underlying allergic diseases for improved patient care.
Collapse
Affiliation(s)
- Xiaorui Han
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University Stanford University Stanford CA USA
| | - James W. Krempski
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University Stanford University Stanford CA USA
| | - Kari Nadeau
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University Stanford University Stanford CA USA
| |
Collapse
|
50
|
Maturation of the gut microbiome during the first year of life contributes to the protective farm effect on childhood asthma. Nat Med 2020; 26:1766-1775. [PMID: 33139948 DOI: 10.1038/s41591-020-1095-x] [Citation(s) in RCA: 182] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 09/08/2020] [Indexed: 12/19/2022]
Abstract
Growing up on a farm is associated with an asthma-protective effect, but the mechanisms underlying this effect are largely unknown. In the Protection against Allergy: Study in Rural Environments (PASTURE) birth cohort, we modeled maturation using 16S rRNA sequence data of the human gut microbiome in infants from 2 to 12 months of age. The estimated microbiome age (EMA) in 12-month-old infants was associated with previous farm exposure (β = 0.27 (0.12-0.43), P = 0.001, n = 618) and reduced risk of asthma at school age (odds ratio (OR) = 0.72 (0.56-0.93), P = 0.011). EMA mediated the protective farm effect by 19%. In a nested case-control sample (n = 138), we found inverse associations of asthma with the measured level of fecal butyrate (OR = 0.28 (0.09-0.91), P = 0.034), bacterial taxa that predict butyrate production (OR = 0.38 (0.17-0.84), P = 0.017) and the relative abundance of the gene encoding butyryl-coenzyme A (CoA):acetate-CoA-transferase, a major enzyme in butyrate metabolism (OR = 0.43 (0.19-0.97), P = 0.042). The gut microbiome may contribute to asthma protection through metabolites, supporting the concept of a gut-lung axis in humans.
Collapse
|