1
|
Saeki H, Ohya Y, Arakawa H, Ichiyama S, Katsunuma T, Katoh N, Tanaka A, Tanizaki H, Tsunemi Y, Nakahara T, Nagao M, Narita M, Hide M, Fujisawa T, Futamura M, Masuda K, Matsubara T, Murota H, Yamamoto-Hanada K, Furuta J. English version of clinical practice guidelines for the management of atopic dermatitis 2024. J Dermatol 2025; 52:e70-e142. [PMID: 39707640 DOI: 10.1111/1346-8138.17544] [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: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 12/23/2024]
Abstract
This is the English version of the 2024 clinical practice guidelines for the management of atopic dermatitis (AD). AD is a disease characterized by relapsing eczema with pruritus as a primary lesion. A crucial aspect of AD treatment is the prompt induction of remission via the suppression of existing skin inflammation and pruritus. To achieve this, topical anti-inflammatory drugs, such as topical corticosteroids, tacrolimus ointment, delgocitinib ointment, and difamilast ointment, have been used. However, the following treatments should be considered in addition to topical therapy for patients with refractory moderate-to-severe AD: oral cyclosporine, subcutaneous injections of biologics (dupilumab, nemolizumab, tralokinumab), oral Janus kinase inhibitors (baricitinib, upadacitinib, abrocitinib), and phototherapy. In these revised guidelines, descriptions of five new drugs, namely, difamilast, nemolizumab, tralokinumab, upadacitinib, and abrocitinib, have been added. The guidelines present recommendations to review clinical research articles, evaluate the balance between the advantages and disadvantages of medical activities, and optimize medical activity-related patient outcomes with respect to several important points requiring decision-making in clinical practice.
Collapse
MESH Headings
- Humans
- Dermatitis, Atopic/drug therapy
- Dermatitis, Atopic/diagnosis
- Dermatitis, Atopic/therapy
- Janus Kinase Inhibitors/therapeutic use
- Dermatologic Agents/therapeutic use
- Dermatologic Agents/administration & dosage
- Administration, Cutaneous
- Severity of Illness Index
- Administration, Oral
- Injections, Subcutaneous
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal
Collapse
Affiliation(s)
- Hidehisa Saeki
- Department of Dermatology, Nippon Medical School, Tokyo, Japan
| | - Yukihiro Ohya
- Department of Occupational and Environmental Health, Graduate School of Medical Sciences and Medical School, Nagoya City University, Nagoya, Japan
| | - Hirokazu Arakawa
- Kitakanto Allergy Research Institute, Kibounoie Hospital, Gunma, Japan
| | - Susumu Ichiyama
- Department of Dermatology, Nippon Medical School, Tokyo, Japan
| | - Toshio Katsunuma
- Department of Pediatrics, The Jikei University Daisan Hospital, Tokyo, Japan
| | - Norito Katoh
- Department for Medical Innovation and Translational Medical Science, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Akio Tanaka
- Department of Dermatology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hideaki Tanizaki
- Department of Dermatology, Kansai Medical University, Osaka, Japan
| | - Yuichiro Tsunemi
- Department of Dermatology, Saitama Medical University, Saitama, Japan
| | - Takeshi Nakahara
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Mizuho Nagao
- Allergy Center, National Hospital Organization Mie National Hospital, Tsu, Japan
| | - Masami Narita
- Department of Pediatrics, Faculty of Medicine, Kyorin University, Tokyo, Japan
| | - Michihiro Hide
- Department of Dermatology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Department of Dermatology, Hiroshima City Hiroshima Citizens Hospital, Hiroshima, Japan
| | - Takao Fujisawa
- Allergy Center, National Hospital Organization Mie National Hospital, Tsu, Japan
| | - Masaki Futamura
- Division of Pediatrics, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Koji Masuda
- Department of Dermatology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Tomoyo Matsubara
- Department of Pediatrics, Dokkyo Medical University Saitama Medical Center, Saitama, Japan
| | - Hiroyuki Murota
- Department of Dermatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | | | - Junichi Furuta
- Medical Informatics and Management, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
2
|
Das A, Fleming P. Evaluating probiotic mechanisms may help inform optimum strain selection for use in clinical trials. Pediatr Res 2024; 96:1537-1538. [PMID: 39014243 DOI: 10.1038/s41390-024-03395-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/30/2024] [Accepted: 05/22/2024] [Indexed: 07/18/2024]
Affiliation(s)
- Abhishek Das
- Division of Infection and Immunity, University College London, London, WC1E 6BT, UK
| | - Paul Fleming
- Department of Neonatology, Homerton Healthcare NHS Foundation Trust, London, UK.
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
3
|
Balan D, Baral T, Manu MK, Mohapatra AK, Miraj SS. Efficacy of probiotics as adjuvant therapy in bronchial asthma: a systematic review and meta-analysis. ALLERGY, ASTHMA, AND CLINICAL IMMUNOLOGY : OFFICIAL JOURNAL OF THE CANADIAN SOCIETY OF ALLERGY AND CLINICAL IMMUNOLOGY 2024; 20:60. [PMID: 39563347 PMCID: PMC11575415 DOI: 10.1186/s13223-024-00922-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 10/31/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND Asthma is a chronic, heterogeneous disease characterized by airway inflammation. Asthma exacerbations significantly increase the disease burden, necessitating new therapeutic approaches. Emerging evidence suggests probiotics, through the gut-lung axis, may benefit asthma management by modulating immune responses and reducing inflammation. METHODS This systematic review and meta-analysis adhered to PRISMA guidelines and was registered with PROSPERO (CRD42023480098). A comprehensive search of PubMed, Scopus, Web of Science, and Embase was conducted up to March 2024. Inclusion criteria encompassed randomized controlled trials (RCTs) evaluating probiotic interventions in asthma patients. Statistical analysis was done using RevMan 5.3, with odds ratios (OR) and 95% confidence intervals (CI) calculated, and heterogeneity assessed using I2 statistics. RESULTS Twelve RCTs, comprising 1401 participants, met the inclusion criteria. The probiotic strains investigated included various Lactobacillus and Bifidobacterium species. Meta-analysis revealed significant improvements in asthma control test scores (OR 1.18, 95% CI: 1.18-3.64, p = 0.0001) following probiotic supplementation. Probiotics also improved fractional exhaled nitric oxide (FeNO) in one study, but pooled FeNO and eosinophil data were not statistically significant (p = 0.46 and p = 0.29, respectively). One study observed fewer asthma exacerbations in the probiotic group (24/212) compared to placebo (67/210), with no difference in exacerbation duration. CONCLUSION Probiotic supplementation may be beneficial in improving asthma symptom control with no significant impact on lung function indices or eosinophil levels. Probiotics can be a potential adjunctive therapy in asthma management, particularly for asthma symptom control.
Collapse
Affiliation(s)
- Divya Balan
- Department of Respiratory Medicine, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Tejaswini Baral
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Mohan K Manu
- Department of Respiratory Medicine, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| | - Aswini Kumar Mohapatra
- Department of Respiratory Medicine, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Sonal Sekhar Miraj
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| |
Collapse
|
4
|
Zhong B, Liang W, Zhao Y, Li F, Zhao Z, Gao Y, Yang G, Li S. Combination of Lactiplantibacillus Plantarum ELF051 and Astragalus Polysaccharides Improves Intestinal Barrier Function and Gut Microbiota Profiles in Mice with Antibiotic-Associated Diarrhea. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10368-3. [PMID: 39354215 DOI: 10.1007/s12602-024-10368-3] [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] [Accepted: 09/17/2024] [Indexed: 10/03/2024]
Abstract
The purpose of this study was to investigate the improvement of the intestinal barrier and gut microbiota in mice with antibiotic-associated diarrhea (AAD) using Lactiplantibacillus plantarum ELF051 combined with Astragalus polysaccharides. The amoxicillin, clindamycin, and streptomycin triple-mixed antibiotic-induced AAD models were administered with L. plantarum ELF051 or Astragalus polysaccharides or L. plantarum ELF051 + Astragalus polysaccharides for 14 days. Our findings revealed that the combination of L. plantarum ELF051 and Astragalus polysaccharides elevated the number of goblet cells and enhanced the proportion of mucous within the colon tissue. Furthermore, the expression of sIgA and IgG were upregulated, while the levels of IL-17A, IL-4, DAO, D-LA, LPS, and TGF-β1 were downregulated. L. plantarum ELF051 combined with Astragalus polysaccharides elevated the expression of tight junction (TJ) proteins, facilitating intestinal mucosal repair via Smad signaling nodes. Furthermore, their combination effectively increased the relative abundance of lactic acid bacteria (LAB) and Allobaculum, and decreased the relative abundance of Bacteroides and Blautia. Spearman rank correlation analysis demonstrated that LAB were closely related to permeability factors, immune factors, and indicators of intestinal barrier function. In summary, the effect of combining L. plantarum ELF051 and Astragalus polysaccharides on AAD mice was achieved by enhancing intestinal barrier function and regulating the composition of the gut microbiota.
Collapse
Affiliation(s)
- Bao Zhong
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences (Northeast Agricultural Research Center of China), Changchun, 130033, P.R. China
- College of Food Science and Nutritional Engineering, Jilin Agriculture Science and Technology University, Jilin, 132101, P.R. China
- Brewing Technology Innovation Center of Jilin Province, Jilin Agriculture Science and Technology University, Jilin, 132101, P.R. China
| | - Wei Liang
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences (Northeast Agricultural Research Center of China), Changchun, 130033, P.R. China
- Anshan Hospital of Traditional Chinese Medicine, Anshan, 114004, P.R. China
| | - Yujuan Zhao
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences (Northeast Agricultural Research Center of China), Changchun, 130033, P.R. China
| | - Fenglin Li
- College of Food Science and Nutritional Engineering, Jilin Agriculture Science and Technology University, Jilin, 132101, P.R. China
- Brewing Technology Innovation Center of Jilin Province, Jilin Agriculture Science and Technology University, Jilin, 132101, P.R. China
| | - Zijian Zhao
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences (Northeast Agricultural Research Center of China), Changchun, 130033, P.R. China
| | - Yansong Gao
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences (Northeast Agricultural Research Center of China), Changchun, 130033, P.R. China
| | - Ge Yang
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences (Northeast Agricultural Research Center of China), Changchun, 130033, P.R. China
| | - Shengyu Li
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences (Northeast Agricultural Research Center of China), Changchun, 130033, P.R. China.
| |
Collapse
|
5
|
Gonia S, Heisel T, Miller N, Haapala J, Harnack L, Georgieff MK, Fields DA, Knights D, Jacobs K, Seburg E, Demerath EW, Gale CA, Swanson MH. Maternal oral probiotic use is associated with decreased breastmilk inflammatory markers, infant fecal microbiome variation, and altered recognition memory responses in infants-a pilot observational study. Front Nutr 2024; 11:1456111. [PMID: 39385777 PMCID: PMC11462058 DOI: 10.3389/fnut.2024.1456111] [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: 06/28/2024] [Accepted: 09/02/2024] [Indexed: 10/12/2024] Open
Abstract
Introduction Early life gut microbiomes are important for brain and immune system development in animal models. Probiotic use has been proposed as a strategy to promote health via modulation of microbiomes. In this observational study, we explore if early life exposure to probiotics via the mother during pregnancy and lactation, is associated with decreased inflammation in breastmilk, maternal and infant microbiome variation, and altered infant neurodevelopmental features. Methods Exclusively breastfeeding mother-infant dyads were recruited as part of the "Mothers and Infants Linked for Healthy Growth (MILk) Study." Probiotic comparison groups were defined by exposure to maternal probiotics (NO/YES) and by timing of probiotic exposure (prenatal, postnatal, total). C-reactive protein (CRP) and IL-6 levels were determined in breastmilk by immunoassays, and microbiomes were characterized from 1-month milk and from 1- and 6-month infant feces by 16S rDNA sequencing. Infant brain function was profiled via electroencephalogram (EEG); we assessed recognition memory using event-related potential (ERP) responses to familiar and novel auditory (1 month) and visual (6 months) stimuli. Statistical comparisons of study outcomes between probiotic groups were performed using permutational analysis of variance (PERMANOVA) (microbiome) and linear models (all other study outcomes), including relevant covariables as indicated. Results We observed associations between probiotic exposure and lower breastmilk CRP and IL-6 levels, and infant gut microbiome variation at 1- and 6-months of age (including higher abundances of Bifidobacteria and Lactobacillus). In addition, maternal probiotic exposure was associated with differences in infant ERP features at 6-months of age. Specifically, infants who were exposed to postnatal maternal probiotics (between the 1- and 6-month study visits) via breastfeeding/breastmilk, had larger differential responses between familiar and novel visual stimuli with respect to the late slow wave component of the EEG, which may indicate greater memory updating potential. The milk of mothers of this subgroup of infants had lower IL-6 levels and infants had different 6-month fecal microbiomes as compared to those in the "NO" maternal probiotics group. Discussion These results support continued research into "Microbiota-Gut-Brain" connections during early life and the role of pre- and postnatal probiotics in mothers to promote healthy microbiome-associated outcomes in infants.
Collapse
Affiliation(s)
- Sara Gonia
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
| | - Timothy Heisel
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
| | - Neely Miller
- Department of Psychology, University of Minnesota, Minneapolis, MN, United States
| | - Jacob Haapala
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, United States
| | - Lisa Harnack
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, United States
| | - Michael K. Georgieff
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
| | - David A. Fields
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Dan Knights
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Katherine Jacobs
- Division of Maternal-Fetal Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Elisabeth Seburg
- Pregnancy and Child Health Research Center, HealthPartners Institute, Bloomington, MN, United States
| | - Ellen W. Demerath
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, United States
| | - Cheryl A. Gale
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
| | - Marie H. Swanson
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
6
|
Wang H, He Y, Dang D, Feng L, Huang L, Zhao J, Lu S, Lu W. Bifidobacterium animalis subsp. lactis CCFM1274 relieved allergic asthma symptoms by modifying intestinal tryptophan metabolism in mice. Food Funct 2024; 15:8810-8822. [PMID: 39115430 DOI: 10.1039/d4fo01079e] [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: 08/28/2024]
Abstract
Approximately two-thirds of patients with asthma, a common inflammatory airway disease, are thought to present with allergies. Probiotics and tryptophan metabolites are becoming increasingly important in treating allergic asthma. This study aimed to identify potential probiotic strains and tryptophan metabolites that could alleviate asthma symptoms. Based on in vitro fermentation experiments, we evaluated variations in probiotic capacity to metabolize tryptophan. Of the eight tested strains, Bifidobacterium animalis subsp. lactis CCFM1274 produced relatively high levels of indole-3-carboxaldehyde (I3C). A mouse model of allergic asthma was established by oral administration of ovalbumin (OVA) and was subjected to oral administration of probiotics. The results demonstrated that treatment with CCFM1274 reduced the tendency for body weight loss and mortality in OVA-induced asthmatic mice. Ingestion of CCFM1274 improved the infiltration of perivascular and peribronchial inflammatory cells in the lung sections stained with hematoxylin and eosin (H&E). This outcome was accompanied by a reduction in the serum levels of OVA-specific immunoglobulin E (OVA-sIgE) and in the levels of IL-10 and IL-17 in the bronchoalveolar lavage fluid (BALF). The linear discriminant analysis effect size (LEfSe) of the gut microbiota showed that CCFM1274 increased the relative abundance of Bifidobacterium. In conclusion, CCFM1274 remodeled intestinal tryptophan metabolism in mice and contributed to the improvement of allergic asthma.
Collapse
Affiliation(s)
- Hongchao Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yuan He
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Danting Dang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Ling Feng
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Liming Huang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Shourong Lu
- Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu 214023, China.
| | - Wenwei Lu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| |
Collapse
|
7
|
Puisto R, Gómez-Gallego C, Collado MC, Turta O, Isolauri E, Rautava S. The Role of Infant Gut Microbiota Modulation by Perinatal Maternal Probiotic Intervention in Atopic Eczema Risk Reduction. Neonatology 2024:1-11. [PMID: 39074448 DOI: 10.1159/000540075] [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: 02/10/2024] [Accepted: 06/24/2024] [Indexed: 07/31/2024]
Abstract
INTRODUCTION Probiotics have shown potential in reducing the occurrence of atopic eczema in high-risk infants. We aimed here to assess whether the preventive effect of maternal probiotic administration stems from compositional changes in early gut microbiota. METHODS This study included 46 mother-infant pairs from an original randomized controlled trial assessing the impact of maternal probiotic intervention with either the combinations of Lacticaseibacillus rhamnosus LPR and Bifidobacterium longum BL999, or Lacticaseibacillus paracasei ST11 and Bifidobacterium longum BL999, or placebo beginning 2 months before expected delivery and ending 2 months after birth. All children were vaginally delivered, full term and breastfed. During the 2-year follow-up period, the children were clinically evaluated by physicians for atopic eczema, and their gut microbiota was profiled at 1 and 6 months of age by 16S rRNA gene sequencing using an Illumina sequencing platform. RESULTS Altogether, 19 of 46 children developed atopic eczema by the age of 2 years. At 1 and 6 months of age, gut microbial diversity was similar between children who developed atopic eczema and their healthy controls, but at the age of 6 months, children who developed atopic eczema manifested with significantly higher relative abundance of Clostridia. Probiotic intervention did not significantly influence microbial diversity, and the effects on microbial composition were not consistent with the changes associated with the development of atopic eczema. CONCLUSION The reduction of the risk of atopic eczema achieved by perinatal maternal probiotic intervention does not seem to require substantial gut microbiota modulation.
Collapse
Affiliation(s)
- Reetta Puisto
- Department of Pediatrics, Faculty of Medicine, University of Turku, Turku, Finland
| | - Carlos Gómez-Gallego
- Institute of Public Health and Clinical Nutrition, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Olli Turta
- Department of Pediatrics, Faculty of Medicine, University of Turku, Turku, Finland
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
| | - Erika Isolauri
- Department of Pediatrics, Faculty of Medicine, University of Turku, Turku, Finland
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
| | - Samuli Rautava
- Department of Pediatrics, Faculty of Medicine, University of Turku, Turku, Finland
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
| |
Collapse
|
8
|
Morita A, Ichihara E, Inoue K, Fujiwara K, Yokoyama T, Harada D, Ando C, Kano H, Oda N, Tamura T, Ochi N, Kawai H, Inoue M, Hara N, Fujimoto N, Ichikawa H, Oze I, Hotta K, Maeda Y, Kiura K. Impacts of probiotics on the efficacies of immune checkpoint inhibitors with or without chemotherapy for patients with advanced non-small-cell lung cancer. Int J Cancer 2024; 154:1607-1615. [PMID: 38196128 DOI: 10.1002/ijc.34842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 12/08/2023] [Accepted: 12/11/2023] [Indexed: 01/11/2024]
Abstract
The relationships between the therapeutic effects of immune checkpoint inhibitors (ICIs) and the intestinal flora have attracted increasing attention. However, the effects of oral probiotics on the efficacies of ICIs used to treat non-small-cell lung cancer (NSCLC) remain unclear. We investigated the effects of probiotics on the efficacies of ICIs in patients treated with and without chemotherapy. We investigated patients with advanced NSCLC on ICI monotherapy or combination ICI and chemotherapy using the Okayama Lung Cancer Study Group Immunotherapy Database (OLCSG-ID) and the Okayama Lung Cancer Study Group Immunochemotherapy Database (OLCSG-ICD). In total, 927 patients (482 on ICI monotherapy, 445 on an ICI + chemotherapy) were enrolled. Most were male, of good performance status, smokers, and without epidermal growth factor receptor (EGFR)/anaplastic lymphoma kinase (ALK) mutations. Probiotics were administered to 19% of patients on ICI monotherapies and 17% of those on ICIs + chemotherapy. Of the former patients, progression-free survival (PFS) and overall survival (OS) were significantly better in the probiotics group (PFS 7.9 vs. 2.9 months, hazard ratio [HR] 0.54, p < .001; OS not attained vs. 13.1 months, HR 0.45, p < .001). Among patients receiving ICI and chemotherapy, there were no significant differences in PFS between those on probiotics and not but OS was significantly better in the probiotics group (PFS 8.8 vs. 8.6 months, HR 0.89, p = .43; OS not attained vs. 22.6 months, HR 0.61, p = .03). Patients on probiotics experienced better outcomes following ICI treatment.
Collapse
Affiliation(s)
- Ayako Morita
- Department of Hematology and Oncology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Eiki Ichihara
- Center for Clinical Oncology, Okayama University Hospital, Okayama, Japan
| | - Koji Inoue
- Department of Respiratory Medicine, Ehime Prefectural Central Hospital, Matsuyama, Japan
| | - Keiichi Fujiwara
- Department of Respiratory Medicine, NHO Okayama Medical Center, Okayama, Japan
| | - Toshihide Yokoyama
- Department of Respiratory Medicine, Ohara Healthcare Foundation, Kurashiki Central Hospital, Kurashiki, Japan
| | - Daijiro Harada
- Department of Thoracic Oncology, NHO Shikoku Cancer Center, Matsuyama, Japan
| | - Chihiro Ando
- Department of Respiratory Medicine, Japanese Red Cross Okayama Hospital, Okayama, Japan
| | - Hirohisa Kano
- Department of Respiratory Medicine, Japanese Red Cross Okayama Hospital, Okayama, Japan
| | - Naohiro Oda
- Department of Respiratory Medicine, Fukuyama City Hospital, Fukuyama, Japan
| | - Tomoki Tamura
- Department of Respiratory Medicine, NHO Iwakuni Clinical Center, Iwakuni, Japan
| | - Nobuaki Ochi
- Department of General Internal Medicine 4, Kawasaki Medical School, Okayama, Japan
| | - Haruyuki Kawai
- Department of Internal Medicine, Okayama Saiseikai General Hospital, Okayama, Japan
| | - Masaaki Inoue
- Department of Chest Surgery, Shimonoseki City Hospital, Shimonoseki, Japan
| | - Naofumi Hara
- Department of Respiratory Medicine, Okayama Rosai Hospital, Okayama, Japan
| | - Nobukazu Fujimoto
- Department of Respiratory Medicine, Okayama Rosai Hospital, Okayama, Japan
| | - Hirohisa Ichikawa
- Department of Respiratory Medicine, KKR Takamatsu Hospital, Takamatsu, Japan
| | - Isao Oze
- Division of Cancer Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Katsuyuki Hotta
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama, Japan
| | - Yoshinobu Maeda
- Department of Hematology and Oncology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Katsuyuki Kiura
- Department of Allergy and Respiratory Medicine, Okayama University Hospital, Okayama, Japan
| |
Collapse
|
9
|
Das S, Konwar BK. Influence of connatural factors in shaping vaginal microflora and ensuring its health. Arch Gynecol Obstet 2024; 309:871-886. [PMID: 37676318 DOI: 10.1007/s00404-023-07200-8] [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: 06/12/2022] [Accepted: 08/21/2023] [Indexed: 09/08/2023]
Abstract
Vaginal canal (VC) is exposed to the external environment affected by habitual factors like hygiene and sexual behaviour as well as physiological factors like puberty, menstrual cycle, pregnancy, child birth and menopause. Healthy VC harbours beneficial microflora supported by vaginal epithelium and cervical fluid. Connatural antimicrobial peptide (AMPs) of female reproductive tract (FRT) conjunctly with these beneficial microbes provide protection from a large number of infectious diseases. Such infections may either be caused by native microbes of the VC or transitory microbes like bacteria or virus which are not a part of VC microflora. This review highlight's the role of hormones, enzymes, innate immunological factors, epithelial cells and vaginal mucus that support beneficial microbes over infectious ones thus, helping to maintain homeostasis in VC and further protect the FRT. We also discuss the prospective use of vaginal probiotics and AMPs against pathogens which can serve as a potential cure for vaginal infections.
Collapse
Affiliation(s)
- Shreaya Das
- Department of MBBT, Tezpur University, Napaam, Assam, 784028, India.
| | - Bolin K Konwar
- Department of MBBT, Tezpur University, Napaam, Assam, 784028, India
| |
Collapse
|
10
|
Milani TMS, Sandy CM, Calazans APCT, Silva RQ, Fonseca VMB, Martins FS, Borges MC. Dose-Response Effect of Saccharomyces cerevisiae UFMG A-905 on the Prevention of Asthma in an Animal Model. Probiotics Antimicrob Proteins 2024; 16:53-61. [PMID: 36445686 DOI: 10.1007/s12602-022-10014-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2022] [Indexed: 11/30/2022]
Abstract
Probiotics should be administered in adequate amounts to confer health benefits. Probiotic dose-response studies are still missing. Saccharomyces cerevisiae UFMG A-905 prevented asthma development; however, the ideal dose has not been investigated. We evaluated the optimal dose and administration regimen of S. cerevisiae UFMG A-905 in the prevention of asthma. Male Balb/c mice were sensitized intraperitoneally with ovalbumin (OVA) and challenged with OVA intranasally. Mice received, via gavage, daily or alternate-day S. cerevisiae UFMG A-905. In daily regimen, different concentrations (107, 108, or 109 CFU/mL) were given 10 days before OVA sensitization and during challenges. In alternate-day regimen, a concentration of 109 CFU/mL was administered three times per week for 5 weeks, starting 2 weeks prior to the first sensitization. After the last challenge, in vivo bronchial hyperresponsiveness and airway and lung inflammation were assessed. OVA-challenged mice, when compared to saline-challenged mice, presented a significant increase in bronchial hyperresponsiveness and airway and lung inflammation. Daily and alternate-day administration of 109 CFU/mL of S. cerevisiae UFMG A-905 significantly reduced bronchial hyperresponsiveness; lower concentrations of S. cerevisiae UFMG A-905 did not significantly reduce bronchial hyperresponsiveness. Daily regimen with the highest concentration significantly reduced total cell number, eosinophil count in the BAL, and the levels of IL-4, IL-5, and IL-13. Daily administration of S. cerevisiae UFMG A-905 at 107 and 108 CFU/mL and alternate-day regimen did not significantly decrease airway and lung inflammation. S. cerevisiae UFMG A-905 led to a significant attenuation of bronchial hyperresponsiveness and lung inflammation in a dose-dependent manner.
Collapse
Affiliation(s)
- Thamires M S Milani
- Department of Internal Medicine, Ribeirao Preto Medical School, University of São Paulo, São Paulo, SP, Brazil
| | - Camila M Sandy
- Department of Internal Medicine, Ribeirao Preto Medical School, University of São Paulo, São Paulo, SP, Brazil
| | | | - Rosana Q Silva
- Department of Internal Medicine, Ribeirao Preto Medical School, University of São Paulo, São Paulo, SP, Brazil
| | - Vanessa M B Fonseca
- Department of Internal Medicine, Ribeirao Preto Medical School, University of São Paulo, São Paulo, SP, Brazil
| | - Flaviano S Martins
- Department of Microbiology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, São Paulo, MG, Brazil
| | - Marcos C Borges
- Department of Internal Medicine, Ribeirao Preto Medical School, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
11
|
Reuter S, Raspe J, Taube C. Microbes little helpers and suppliers for therapeutic asthma approaches. Respir Res 2024; 25:29. [PMID: 38218816 PMCID: PMC10787474 DOI: 10.1186/s12931-023-02660-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/28/2023] [Indexed: 01/15/2024] Open
Abstract
Bronchial asthma is a prevalent and increasingly chronic inflammatory lung disease affecting over 300 million people globally. Initially considered an allergic disorder driven by mast cells and eosinophils, asthma is now recognized as a complex syndrome with various clinical phenotypes and immunological endotypes. These encompass type 2 inflammatory endotypes characterized by interleukin (IL)-4, IL-5, and IL-13 dominance, alongside others featuring mixed or non-eosinophilic inflammation. Therapeutic success varies significantly based on asthma phenotypes, with inhaled corticosteroids and beta-2 agonists effective for milder forms, but limited in severe cases. Novel antibody-based therapies have shown promise, primarily for severe allergic and type 2-high asthma. To address this gap, novel treatment strategies are essential for better control of asthma pathology, prevention, and exacerbation reduction. One promising approach involves stimulating endogenous anti-inflammatory responses through regulatory T cells (Tregs). Tregs play a vital role in maintaining immune homeostasis, preventing autoimmunity, and mitigating excessive inflammation after pathogenic encounters. Tregs have demonstrated their ability to control both type 2-high and type 2-low inflammation in murine models and dampen human cell-dependent allergic airway inflammation. Furthermore, microbes, typically associated with disease development, have shown immune-dampening properties that could be harnessed for therapeutic benefits. Both commensal microbiota and pathogenic microbes have demonstrated potential in bacterial-host interactions for therapeutic purposes. This review explores microbe-associated approaches as potential treatments for inflammatory diseases, shedding light on current and future therapeutics.
Collapse
Affiliation(s)
- Sebastian Reuter
- Department of Pulmonary Medicine, University Hospital Essen-Ruhrlandklinik, Tüschener Weg 40, 45239, Essen, Germany.
| | - Jonas Raspe
- Department of Pulmonary Medicine, University Hospital Essen-Ruhrlandklinik, Tüschener Weg 40, 45239, Essen, Germany
| | - Christian Taube
- Department of Pulmonary Medicine, University Hospital Essen-Ruhrlandklinik, Tüschener Weg 40, 45239, Essen, Germany
| |
Collapse
|
12
|
Zhang Q, Zhang C, Zhang Y, Liu Y, Wang J, Gao Z, Sun J, Li Q, Sun J, Cui X, Wang Y, Fu L. Early-life risk factors for food allergy: Dietary and environmental factors revisited. Compr Rev Food Sci Food Saf 2023; 22:4355-4377. [PMID: 37679957 DOI: 10.1111/1541-4337.13226] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 07/21/2023] [Accepted: 07/27/2023] [Indexed: 09/09/2023]
Abstract
There appears a steep increase in the prevalence of food allergy worldwide in the past few decades. It is believed that, rather than genetic factors, the recently altered dietary and environmental factors are the driving forces behind the rapid increase of this disease. Accumulating evidence has implied that external exposures that occurred in prenatal and postnatal periods could affect the development of oral tolerance in later life. Understanding the potential risk factors for food allergy would greatly benefit the progress of intervention and therapy. In this review, we present updated knowledge on the dietary and environmental risk factors in early life that have been shown to impact the development of food allergy. These predominantly include dietary habits, microbial exposures, allergen exposure routes, environmental pollutants, and so on. The key evidence, conflicts, and potential research topics of each theory are discussed, and associated interventional strategies to prevent the disease development and ameliorate treatment burden are included. Accumulating evidence has supported the causative role of certain dietary and environmental factors in the establishment of oral tolerance in early life, especially the time of introducing allergenic foods, skin barrier function, and microbial exposures. In addition to certain immunomodulatory factors, increasing interest is raised toward modern dietary patterns, where adequately powered studies are required to identify contributions of those modifiable risk factors. This review broadens our understanding of the connections between diet, environment, and early-life immunity, thus benefiting the progress of intervention and therapy of food allergy.
Collapse
Affiliation(s)
- Qiaozhi Zhang
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Chi Zhang
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Yong Zhang
- Nutrition Department of the First Medical Centre of PLA General Hospital, Beijing, China
| | - Yinghua Liu
- Nutrition Department of the First Medical Centre of PLA General Hospital, Beijing, China
| | - Jin Wang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, China
- Department of Bioresource Engineering, Faculty of Agricultural and Environmental Sciences, McGill University, Sainte-Anne-de-Bellevue, QC, Canada
| | - Zhongshan Gao
- Allergy Research Center, Zhejiang University, Hangzhou, China
| | - Jinlyu Sun
- Allergy Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qianqian Li
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Jiachen Sun
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Xin Cui
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Yanbo Wang
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Linglin Fu
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| |
Collapse
|
13
|
Komulainen M, Saros L, Vahlberg T, Nermes M, Jartti T, Laitinen K. Maternal fish oil and/or probiotics intervention: Allergic diseases in children up to two years old. Pediatr Allergy Immunol 2023; 34:e14004. [PMID: 37622257 DOI: 10.1111/pai.14004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 07/11/2023] [Accepted: 07/25/2023] [Indexed: 08/26/2023]
Abstract
BACKGROUND As n-3 long-chain polyunsaturated fatty acids and probiotics possess immunomodulatory properties, theoretically they could lower the risk of allergic diseases. But their effects remain controversial. We aimed to study the effects of fish oil and probiotics separately or in combination from early pregnancy onwards to lower the risk of allergic diseases in the infants. METHODS In this double-blind trial, women (n = 439) in early pregnancies were randomized into four intervention groups: fish oil + placebo, probiotics + placebo, fish oil + probiotics, and placebo + placebo. Fish oil (1.9 g docosahexaenoic acid and 0.22 g eicosapentaenoic acid) and probiotic (Lacticaseibacillus rhamnosus HN001 and Bifidobacterium animalis ssp. lactis 420, 1010 colony-forming units each) supplements were provided for daily consumption from randomization up to 6 months postpartum. All analyses were adjusted with pet ownership. RESULTS No difference between the infants in the four intervention groups were found regarding physician-diagnosed food allergy, atopic eczema, or atopy at the age of 12 or 24 months (all p > .05). The probiotic intervention was associated with lower odds of recurrent wheezing at 24 months (OR 0.39, 95% CI 0.18-0.84, p = .017), but not at 12 months. CONCLUSIONS The use of fish oil and/or probiotics from early pregnancy onwards did not lower the odds of childhood allergic diseases or atopy, with the exception of the probiotic intervention which decreased the risk of recurrent wheezing when the infants were two years old. This suggests that the incidence of asthma could also decrease later in childhood and thus these outcomes need to be clarified in further investigations.
Collapse
Affiliation(s)
- Miisa Komulainen
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
| | - Lotta Saros
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
| | - Tero Vahlberg
- Department of Clinical Medicine, Biostatistics, University of Turku, Turku, Finland
| | - Merja Nermes
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Tuomas Jartti
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
- Department of Pediatrics and Adolescent Medicine, University of Oulu, Oulu, Finland
| | - Kirsi Laitinen
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
- Functional Foods Forum, University of Turku, Turku, Finland
| |
Collapse
|
14
|
Heldner A, Heath MD, Schnautz B, Kotz S, Chaker A, Kramer MF, Jakwerth CA, Zissler UM, Schmidt-Weber CB, Blank S. Ex Vivo Immunomodulatory Effects of Lactobacillus-, Lacticaseibacillus-, and Bifidobacterium-Containing Synbiotics on Human Peripheral Blood Mononuclear Cells and Monocyte-Derived Dendritic Cells in the Context of Grass Pollen Allergy. Probiotics Antimicrob Proteins 2023; 15:868-879. [PMID: 35113319 PMCID: PMC10393851 DOI: 10.1007/s12602-022-09920-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2022] [Indexed: 11/26/2022]
Abstract
Sensing of the intestinal microbiota by the host immune system is important to induce protective immune responses. Hence, modification of the gut microbiota might be able to prevent or treat allergies, mediated by proinflammatory Th2 immune responses. The aim was to investigate the ex vivo immunomodulatory effects of the synbiotics Pollagen® and Kallergen®, containing the probiotic bacterial strains Lactobacillus, Lacticaseibacillus and Bifidobacterium, in the context of grass pollen allergy. Peripheral blood mononuclear cells (PBMCs) from grass pollen-allergic patients and healthy controls were stimulated with grass pollen extract (GPE) and synbiotics and Gata3 expression and cytokine secretion analyzed. Monocyte-derived dendritic cells (MoDCs) cells were matured in the presence of GPE and synbiotics, co-cultured with autologous naïve T cells and maturation markers and cytokine secretion analyzed. GPE stimulation of PBMCs from grass pollen-allergic patients resulted in a significant higher production of the Th2 cytokines IL-4, IL-5, IL-9 and IL-13 compared to healthy controls. Gata3+CD4+ T cell induction was independent of the allergic status. The synbiotics promoted IL-10 and IFN-γ secretion and downregulated the GPE-induced Th2-like phenotype. Co-culturing naïve T cells with MoDCs, matured in the presence of GPE and synbiotics, shifted the GPE-induced Th2 cytokine release towards Th1-Th17-promoting conditions in allergic subjects. The investigated synbiotics are effective in downregulating the GPE-induced Th2 immune response in PBMCs from grass pollen-allergic patients as well as in autologous MoDC-T cell stimulation assays. In addition to increased IL-10 release, the data indicates a shift from a Th2- to a more Th1- and Th17-like phenotype.
Collapse
Affiliation(s)
- Alexander Heldner
- Center of Allergy and Environment (ZAUM)Faculty of Medicine and Helmholtz Center MunichMember of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Technical University of Munich, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764, Munich, Germany
| | | | - Benjamin Schnautz
- Center of Allergy and Environment (ZAUM)Faculty of Medicine and Helmholtz Center MunichMember of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Technical University of Munich, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764, Munich, Germany
| | - Sebastian Kotz
- Faculty of Medicine, Department of Otolaryngology, Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
| | - Adam Chaker
- Center of Allergy and Environment (ZAUM)Faculty of Medicine and Helmholtz Center MunichMember of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Technical University of Munich, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764, Munich, Germany
- Faculty of Medicine, Department of Otolaryngology, Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
| | - Matthias F Kramer
- Allergy Therapeutics PLC, Worthing, UK
- Bencard Allergie GmbH, Munich, Germany
| | - Constanze A Jakwerth
- Center of Allergy and Environment (ZAUM)Faculty of Medicine and Helmholtz Center MunichMember of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Technical University of Munich, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764, Munich, Germany
| | - Ulrich M Zissler
- Center of Allergy and Environment (ZAUM)Faculty of Medicine and Helmholtz Center MunichMember of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Technical University of Munich, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764, Munich, Germany
| | - Carsten B Schmidt-Weber
- Center of Allergy and Environment (ZAUM)Faculty of Medicine and Helmholtz Center MunichMember of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Technical University of Munich, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764, Munich, Germany
| | - Simon Blank
- Center of Allergy and Environment (ZAUM)Faculty of Medicine and Helmholtz Center MunichMember of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Technical University of Munich, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764, Munich, Germany.
| |
Collapse
|
15
|
Markoulli M, Ahmad S, Arcot J, Arita R, Benitez-Del-Castillo J, Caffery B, Downie LE, Edwards K, Flanagan J, Labetoulle M, Misra SL, Mrugacz M, Singh S, Sheppard J, Vehof J, Versura P, Willcox MDP, Ziemanski J, Wolffsohn JS. TFOS Lifestyle: Impact of nutrition on the ocular surface. Ocul Surf 2023; 29:226-271. [PMID: 37100346 DOI: 10.1016/j.jtos.2023.04.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 04/06/2023] [Indexed: 04/28/2023]
Abstract
Nutrients, required by human bodies to perform life-sustaining functions, are obtained from the diet. They are broadly classified into macronutrients (carbohydrates, lipids, and proteins), micronutrients (vitamins and minerals) and water. All nutrients serve as a source of energy, provide structural support to the body and/or regulate the chemical processes of the body. Food and drinks also consist of non-nutrients that may be beneficial (e.g., antioxidants) or harmful (e.g., dyes or preservatives added to processed foods) to the body and the ocular surface. There is also a complex interplay between systemic disorders and an individual's nutritional status. Changes in the gut microbiome may lead to alterations at the ocular surface. Poor nutrition may exacerbate select systemic conditions. Similarly, certain systemic conditions may affect the uptake, processing and distribution of nutrients by the body. These disorders may lead to deficiencies in micro- and macro-nutrients that are important in maintaining ocular surface health. Medications used to treat these conditions may also cause ocular surface changes. The prevalence of nutrition-related chronic diseases is climbing worldwide. This report sought to review the evidence supporting the impact of nutrition on the ocular surface, either directly or as a consequence of the chronic diseases that result. To address a key question, a systematic review investigated the effects of intentional food restriction on ocular surface health; of the 25 included studies, most investigated Ramadan fasting (56%), followed by bariatric surgery (16%), anorexia nervosa (16%), but none were judged to be of high quality, with no randomized-controlled trials.
Collapse
Affiliation(s)
- Maria Markoulli
- School of Optometry and Vision Science, UNSW Sydney, NSW, Australia.
| | - Sumayya Ahmad
- Icahn School of Medicine of Mt. Sinai, New York, NY, USA
| | - Jayashree Arcot
- Food and Health, School of Chemical Engineering, UNSW Sydney, Australia
| | - Reiko Arita
- Department of Ophthalmology, Itoh Clinic, Saitama, Japan
| | | | | | - Laura E Downie
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Katie Edwards
- School of Optometry and Vision Science, Queensland University of Technology, Brisbane, Australia
| | - Judith Flanagan
- School of Optometry and Vision Science, UNSW Sydney, NSW, Australia; Vision CRC, USA
| | - Marc Labetoulle
- Ophthalmology Department, Hospital Bicêtre, APHP, Paris-Saclay University, Le Kremlin-Bicêtre, France; IDMIT (CEA-Paris Saclay-Inserm U1184), Fontenay-aux-Roses, France
| | - Stuti L Misra
- Department of Ophthalmology, New Zealand National Eye Centre, The University of Auckland, Auckland, New Zealand
| | | | - Sumeer Singh
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - John Sheppard
- Virginia Eye Consultants, Norfolk, VA, USA; Eastern Virginia Medical School, Norfolk, VA, USA
| | - Jelle Vehof
- Departments of Ophthalmology and Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Section of Ophthalmology, School of Life Course Sciences, King's College London, London, UK; Department of Ophthalmology, Vestfold Hospital Trust, Tønsberg, Norway
| | - Piera Versura
- Cornea and Ocular Surface Analysis - Translation Research Laboratory, Ophthalmology Unit, DIMEC Alma Mater Studiorum Università di Bologna, Italy; IRCCS AOU di Bologna Policlinico di Sant'Orsola, Bologna, Italy
| | - Mark D P Willcox
- School of Optometry and Vision Science, UNSW Sydney, NSW, Australia
| | - Jillian Ziemanski
- School of Optometry, University of Alabama at Birmingham, Birmingham, AL, USA
| | - James S Wolffsohn
- College of Health & Life Sciences, School of Optometry, Aston University, Birmingham, UK
| |
Collapse
|
16
|
Wise SK, Damask C, Roland LT, Ebert C, Levy JM, Lin S, Luong A, Rodriguez K, Sedaghat AR, Toskala E, Villwock J, Abdullah B, Akdis C, Alt JA, Ansotegui IJ, Azar A, Baroody F, Benninger MS, Bernstein J, Brook C, Campbell R, Casale T, Chaaban MR, Chew FT, Chambliss J, Cianferoni A, Custovic A, Davis EM, DelGaudio JM, Ellis AK, Flanagan C, Fokkens WJ, Franzese C, Greenhawt M, Gill A, Halderman A, Hohlfeld JM, Incorvaia C, Joe SA, Joshi S, Kuruvilla ME, Kim J, Klein AM, Krouse HJ, Kuan EC, Lang D, Larenas-Linnemann D, Laury AM, Lechner M, Lee SE, Lee VS, Loftus P, Marcus S, Marzouk H, Mattos J, McCoul E, Melen E, Mims JW, Mullol J, Nayak JV, Oppenheimer J, Orlandi RR, Phillips K, Platt M, Ramanathan M, Raymond M, Rhee CS, Reitsma S, Ryan M, Sastre J, Schlosser RJ, Schuman TA, Shaker MS, Sheikh A, Smith KA, Soyka MB, Takashima M, Tang M, Tantilipikorn P, Taw MB, Tversky J, Tyler MA, Veling MC, Wallace D, Wang DY, White A, Zhang L. International consensus statement on allergy and rhinology: Allergic rhinitis - 2023. Int Forum Allergy Rhinol 2023; 13:293-859. [PMID: 36878860 DOI: 10.1002/alr.23090] [Citation(s) in RCA: 139] [Impact Index Per Article: 69.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/11/2022] [Accepted: 09/13/2022] [Indexed: 03/08/2023]
Abstract
BACKGROUND In the 5 years that have passed since the publication of the 2018 International Consensus Statement on Allergy and Rhinology: Allergic Rhinitis (ICAR-Allergic Rhinitis 2018), the literature has expanded substantially. The ICAR-Allergic Rhinitis 2023 update presents 144 individual topics on allergic rhinitis (AR), expanded by over 40 topics from the 2018 document. Originally presented topics from 2018 have also been reviewed and updated. The executive summary highlights key evidence-based findings and recommendation from the full document. METHODS ICAR-Allergic Rhinitis 2023 employed established evidence-based review with recommendation (EBRR) methodology to individually evaluate each topic. Stepwise iterative peer review and consensus was performed for each topic. The final document was then collated and includes the results of this work. RESULTS ICAR-Allergic Rhinitis 2023 includes 10 major content areas and 144 individual topics related to AR. For a substantial proportion of topics included, an aggregate grade of evidence is presented, which is determined by collating the levels of evidence for each available study identified in the literature. For topics in which a diagnostic or therapeutic intervention is considered, a recommendation summary is presented, which considers the aggregate grade of evidence, benefit, harm, and cost. CONCLUSION The ICAR-Allergic Rhinitis 2023 update provides a comprehensive evaluation of AR and the currently available evidence. It is this evidence that contributes to our current knowledge base and recommendations for patient evaluation and treatment.
Collapse
Affiliation(s)
- Sarah K Wise
- Otolaryngology-HNS, Emory University, Atlanta, Georgia, USA
| | - Cecelia Damask
- Otolaryngology-HNS, Private Practice, University of Central Florida, Lake Mary, Florida, USA
| | - Lauren T Roland
- Otolaryngology-HNS, Washington University, St. Louis, Missouri, USA
| | - Charles Ebert
- Otolaryngology-HNS, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Joshua M Levy
- Otolaryngology-HNS, Emory University, Atlanta, Georgia, USA
| | - Sandra Lin
- Otolaryngology-HNS, University of Wisconsin, Madison, Wisconsin, USA
| | - Amber Luong
- Otolaryngology-HNS, McGovern Medical School of the University of Texas, Houston, Texas, USA
| | - Kenneth Rodriguez
- Otolaryngology-HNS, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Ahmad R Sedaghat
- Otolaryngology-HNS, University of Cincinnati, Cincinnati, Ohio, USA
| | - Elina Toskala
- Otolaryngology-HNS, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | - Baharudin Abdullah
- Otolaryngology-HNS, Universiti Sains Malaysia, Kubang, Kerian, Kelantan, Malaysia
| | - Cezmi Akdis
- Immunology, Infectious Diseases, Swiss Institute of Allergy and Asthma Research, Davos, Switzerland
| | - Jeremiah A Alt
- Otolaryngology-HNS, University of Utah, Salt Lake City, Utah, USA
| | | | - Antoine Azar
- Allergy/Immunology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Fuad Baroody
- Otolaryngology-HNS, University of Chicago, Chicago, Illinois, USA
| | | | | | - Christopher Brook
- Otolaryngology-HNS, Harvard University, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Raewyn Campbell
- Otolaryngology-HNS, Macquarie University, Sydney, NSW, Australia
| | - Thomas Casale
- Allergy/Immunology, University of South Florida College of Medicine, Tampa, Florida, USA
| | - Mohamad R Chaaban
- Otolaryngology-HNS, Cleveland Clinic, Case Western Reserve University, Cleveland, Ohio, USA
| | - Fook Tim Chew
- Allergy/Immunology, Genetics, National University of Singapore, Singapore, Singapore
| | - Jeffrey Chambliss
- Allergy/Immunology, University of Texas Southwestern, Dallas, Texas, USA
| | - Antonella Cianferoni
- Allergy/Immunology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | | | | | | | - Anne K Ellis
- Allergy/Immunology, Queens University, Kingston, ON, Canada
| | | | - Wytske J Fokkens
- Otorhinolaryngology, Amsterdam University Medical Centres, Amsterdam, Netherlands
| | | | - Matthew Greenhawt
- Allergy/Immunology, Pediatrics, University of Colorado, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Amarbir Gill
- Otolaryngology-HNS, University of Michigan, Ann Arbor, Michigan, USA
| | - Ashleigh Halderman
- Otolaryngology-HNS, University of Texas Southwestern, Dallas, Texas, USA
| | - Jens M Hohlfeld
- Respiratory Medicine, Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Hannover Medical School, German Center for Lung Research, Hannover, Germany
| | | | - Stephanie A Joe
- Otolaryngology-HNS, University of Illinois Chicago, Chicago, Illinois, USA
| | - Shyam Joshi
- Allergy/Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | | | - Jean Kim
- Otolaryngology-HNS, Johns Hopkins University, Baltimore, Maryland, USA
| | - Adam M Klein
- Otolaryngology-HNS, Emory University, Atlanta, Georgia, USA
| | - Helene J Krouse
- Otorhinolaryngology Nursing, University of Texas Rio Grande Valley, Edinburg, Texas, USA
| | - Edward C Kuan
- Otolaryngology-HNS, University of California Irvine, Orange, California, USA
| | - David Lang
- Allergy/Immunology, Cleveland Clinic, Cleveland, Ohio, USA
| | | | | | - Matt Lechner
- Otolaryngology-HNS, University College London, Barts Health NHS Trust, London, UK
| | - Stella E Lee
- Otolaryngology-HNS, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Victoria S Lee
- Otolaryngology-HNS, University of Illinois Chicago, Chicago, Illinois, USA
| | - Patricia Loftus
- Otolaryngology-HNS, University of California San Francisco, San Francisco, California, USA
| | - Sonya Marcus
- Otolaryngology-HNS, Stony Brook University, Stony Brook, New York, USA
| | - Haidy Marzouk
- Otolaryngology-HNS, State University of New York Upstate, Syracuse, New York, USA
| | - Jose Mattos
- Otolaryngology-HNS, University of Virginia, Charlottesville, Virginia, USA
| | - Edward McCoul
- Otolaryngology-HNS, Ochsner Clinic, New Orleans, Louisiana, USA
| | - Erik Melen
- Pediatric Allergy, Karolinska Institutet, Stockholm, Sweden
| | - James W Mims
- Otolaryngology-HNS, Wake Forest University, Winston Salem, North Carolina, USA
| | - Joaquim Mullol
- Otorhinolaryngology, Hospital Clinic Barcelona, Barcelona, Spain
| | - Jayakar V Nayak
- Otolaryngology-HNS, Stanford University, Palo Alto, California, USA
| | - John Oppenheimer
- Allergy/Immunology, Rutgers, State University of New Jersey, Newark, New Jersey, USA
| | | | - Katie Phillips
- Otolaryngology-HNS, University of Cincinnati, Cincinnati, Ohio, USA
| | - Michael Platt
- Otolaryngology-HNS, Boston University, Boston, Massachusetts, USA
| | | | | | - Chae-Seo Rhee
- Rhinology/Allergy, Seoul National University Hospital and College of Medicine, Seoul, Korea
| | - Sietze Reitsma
- Otolaryngology-HNS, University of Amsterdam, Amsterdam, Netherlands
| | - Matthew Ryan
- Otolaryngology-HNS, University of Texas Southwestern, Dallas, Texas, USA
| | - Joaquin Sastre
- Allergy, Fundacion Jiminez Diaz, University Autonoma de Madrid, Madrid, Spain
| | - Rodney J Schlosser
- Otolaryngology-HNS, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Theodore A Schuman
- Otolaryngology-HNS, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Marcus S Shaker
- Allergy/Immunology, Dartmouth Geisel School of Medicine, Lebanon, New Hampshire, USA
| | - Aziz Sheikh
- Primary Care, University of Edinburgh, Edinburgh, Scotland
| | - Kristine A Smith
- Otolaryngology-HNS, University of Utah, Salt Lake City, Utah, USA
| | - Michael B Soyka
- Otolaryngology-HNS, University of Zurich, University Hospital of Zurich, Zurich, Switzerland
| | - Masayoshi Takashima
- Otolaryngology-HNS, Houston Methodist Academic Institute, Houston, Texas, USA
| | - Monica Tang
- Allergy/Immunology, University of California San Francisco, San Francisco, California, USA
| | | | - Malcolm B Taw
- Integrative East-West Medicine, University of California Los Angeles, Westlake Village, California, USA
| | - Jody Tversky
- Allergy/Immunology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Matthew A Tyler
- Otolaryngology-HNS, University of Minnesota, Minneapolis, Minnesota, USA
| | - Maria C Veling
- Otolaryngology-HNS, University of Texas Southwestern, Dallas, Texas, USA
| | - Dana Wallace
- Allergy/Immunology, Nova Southeastern University, Ft. Lauderdale, Florida, USA
| | - De Yun Wang
- Otolaryngology-HNS, National University of Singapore, Singapore, Singapore
| | - Andrew White
- Allergy/Immunology, Scripps Clinic, San Diego, California, USA
| | - Luo Zhang
- Otolaryngology-HNS, Beijing Tongren Hospital, Beijing, China
| |
Collapse
|
17
|
Schneider R, Sant'Anna A. Using probiotics in paediatric populations. Paediatr Child Health 2022; 27:482-502. [PMID: 36583073 PMCID: PMC9792287 DOI: 10.1093/pch/pxac087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/25/2022] [Indexed: 12/28/2022] Open
Abstract
This statement defines probiotics and reviews the most recent literature on their use in paediatrics. Many studies have examined the potential benefit of probiotics, but significant variation in the strains and doses of probiotics used, the patient populations studied, and in study design, have led to heterogeneous results. Present evidence suggests that probiotics can decrease mortality and lower incidence of necrotizing enterocolitis in preterm and low birth weight neonates. Probiotics may also be beneficial in reducing feeding intolerance. In infants, probiotics may be considered to reduce symptoms of colic. In older children, probiotics can be considered to prevent antibiotic-associated diarrhea and Clostridium difficile -associated diarrhea. Probiotic supplements used in conjunction with standard therapy can help with Helicobacter pylori eradication and decrease the side effects of treatment. Lactobacillus species can be considered to treat irritable bowel syndrome. Probiotics can also be considered to help prevent atopic dermatitis and eczema. To optimize paediatric policy and practice, large, quality studies are needed to determine what types and combinations of probiotics are most efficacious.
Collapse
Affiliation(s)
- Rilla Schneider
- Canadian Paediatric Society, Nutrition and Gastroenterology Committee, Ottawa, Ontario, Canada
| | - Ana Sant'Anna
- Canadian Paediatric Society, Nutrition and Gastroenterology Committee, Ottawa, Ontario, Canada
| |
Collapse
|
18
|
Buchenauer L, Junge KM, Haange SB, Simon JC, von Bergen M, Hoh AL, Aust G, Zenclussen AC, Stangl GI, Polte T. Glyphosate differentially affects the allergic immune response across generations in mice. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 850:157973. [PMID: 35963408 DOI: 10.1016/j.scitotenv.2022.157973] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/24/2022] [Accepted: 08/07/2022] [Indexed: 06/15/2023]
Abstract
Exposure to environmental pollutants via food, particularly during the prenatal and early postnatal periods, has been linked to adverse effects on the immune system. Among these pollutants, the widely used pesticide glyphosate has been associated with endocrine disruption, autism, and cancer. Occupational high exposure to glyphosate has also been shown to influence immune function and exacerbate allergic asthma. However, there are no studies investigating the effect of a common low-dose glyphosate exposure on the allergic immune response - neither directly nor across generations. We therefore explored the impact of oral low-dose glyphosate exposure (0.5 and 50 mg/kg body weight/day) on airway inflammation in dams (F0) and the offspring (F1 and F2 generations) using a murine multi-generational asthma model. While exposure to 50 mg/kg glyphosate induced a mild eosinophilic infiltration in the bronchoalveolar lavage and TH2 cytokine production in the dams, the F1 offspring developed a reduced immune response after maternal exposure to 0.5 mg/kg glyphosate. In particular, decreased lung inflammation, HDM-specific IgE levels, and asthma-relevant cytokine production were primarily observed in the female F1 offspring. However, not only the TH2 cytokines IL-13 and IL-5 but also the TH17 cytokine IL-17 and TH1 cytokine IFN-γ were reduced indicating a more general immunosuppressive function. Notably, the dampened immune response was no longer observed in the female F2 generation. Furthermore, female F1 offspring showed an increased abundance of bacteria in the gut, which have been associated with probiotic-mediated reduced allergic immune responses. Our results suggest a potential immunosuppressive effect of low-dose maternal glyphosate exposure in the F1 offspring that might be mediated by an altered microbiota composition. Further studies are needed to explore if this type of immune response modulation might also be associated with impairments in immune defense upon infectious diseases or even cancer pathology.
Collapse
Affiliation(s)
- Lisa Buchenauer
- Helmholtz Centre for Environmental Research - UFZ, Department of Environmental Immunology, Leipzig, Germany; Department of Dermatology, Venerology and Allergology, Leipzig University Medical Center, University of Leipzig, Leipzig, Germany
| | - Kristin M Junge
- Helmholtz Centre for Environmental Research - UFZ, Department of Environmental Immunology, Leipzig, Germany
| | - Sven-Bastiaan Haange
- Helmholtz Centre for Environmental Research - UFZ, Department of Molecular Systems Biology, Leipzig, Germany
| | - Jan C Simon
- Department of Dermatology, Venerology and Allergology, Leipzig University Medical Center, University of Leipzig, Leipzig, Germany
| | - Martin von Bergen
- Helmholtz Centre for Environmental Research - UFZ, Department of Molecular Systems Biology, Leipzig, Germany; University of Leipzig, Faculty of Life Sciences, Institute of Biochemistry, Leipzig, Germany; German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, Leipzig, Germany
| | - Anna-Lena Hoh
- Research Laboratories and Clinic of Visceral, Transplantation, Thoracic, and Vascular Surgery, Leipzig University and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Gabriela Aust
- Research Laboratories and Clinic of Visceral, Transplantation, Thoracic, and Vascular Surgery, Leipzig University and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Ana C Zenclussen
- Helmholtz Centre for Environmental Research - UFZ, Department of Environmental Immunology, Leipzig, Germany; Perinatal Immunology, Saxonian Incubator for Clinical Translation (SIKT), Medical Faculty, University Leipzig, 04103 Leipzig, Germany
| | - Gabriele I Stangl
- Institute of Agriculture and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Tobias Polte
- Helmholtz Centre for Environmental Research - UFZ, Department of Environmental Immunology, Leipzig, Germany; Department of Dermatology, Venerology and Allergology, Leipzig University Medical Center, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
19
|
Schneider R, Sant'Anna A. L’utilisation des probiotiques dans la population pédiatrique. Paediatr Child Health 2022; 27:482-502. [PMID: 36583070 PMCID: PMC9792288 DOI: 10.1093/pch/pxac086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/25/2022] [Indexed: 12/28/2022] Open
Abstract
Le présent document de principes définit les probiotiques et fournit une analyse des publications scientifiques les plus récentes sur leur utilisation en pédiatrie. De nombreuses études ont évalué les avantages potentiels des probiotiques, mais en raison des variations importantes dans les souches et les doses utilisées, des populations de patients étudiées et des méthodologies privilégiées, les résultats sont hétérogènes. Selon les données probantes à jour, les probiotiques peuvent réduire le taux de mortalité et l’incidence d’entérocolite nécrosante chez les nouveau-nés prématurés et de petit poids à la naissance. Ils peuvent également être bénéfiques pour réduire l’intolérance alimentaire. Chez les nourrissons, on peut envisager de les utiliser pour limiter les symptômes de coliques, et chez les enfants plus âgés, pour prévenir la diarrhée associée aux antibiotiques ou au Clostridium difficile . Les suppléments de probiotiques utilisés conjointement avec un traitement standard peuvent contribuer à éradiquer l’Helicobacter pylori et à atténuer les effets secondaires du traitement. On peut envisager d’utiliser des espèces de Lactobacillus pour traiter le syndrome du côlon irritable ou de recourir à des probiotiques pour contribuer à prévenir la dermatite atopique et l’eczéma. Afin d’optimiser les politiques et les pratiques en pédiatrie, de vastes études de qualité devront être réalisées pour déterminer les types et les combinaisons de probiotiques les plus efficaces.
Collapse
Affiliation(s)
- Rilla Schneider
- Société canadienne de pédiatrie, comité de nutrition et de gastroentérologie, Ottawa (Ontario)Canada
| | - Ana Sant'Anna
- Société canadienne de pédiatrie, comité de nutrition et de gastroentérologie, Ottawa (Ontario)Canada
| |
Collapse
|
20
|
D'Auria E, Acunzo M, Salvatore S, Grazi R, Agosti M, Vandenplas Y, Zuccotti G. Biotics in atopic diseases: state of the art and future perspectives. Minerva Pediatr (Torino) 2022; 74:688-702. [PMID: 36149096 DOI: 10.23736/s2724-5276.22.07010-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Prevalence of allergic diseases has growing in recent decades, being a significant burden for patients and their families. Different environmental factors, acting in early life, can significantly affect the timing and diversity of bacterial colonization and the immune system development. Growing evidence points to a correlation between early life microbial perturbation and development of allergic diseases. Besides, changes in the microbiota in one body site may influence other microbiota communities at distance by different mechanisms, including microbial-derived metabolites, mainly the short chain fatty acids (SCFA). Hence, there has been an increasing interest on the role of "biotics" (probiotics, prebiotics, symbiotics and postbiotics) in shaping dysbiosis and modulating allergic risk. Systemic type 2 inflammation is emerging as a common pathogenetic pathway of allergic diseases, intertwining communication with the gut mcirobiota. The aim of this review was to provide an update overview of the current knowledge of biotics in prevention and treatment of allergic diseases, also addressing research gaps which need to be filled.
Collapse
Affiliation(s)
- Enza D'Auria
- Department of Pediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy -
| | - Miriam Acunzo
- Department of Pediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy
| | - Silvia Salvatore
- Department of Pediatrics, University of Insubria, F. Del Ponte Hospital, Varese, Italy
| | - Roberta Grazi
- Department of Pediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy
| | - Massimo Agosti
- Department of Pediatrics, University of Insubria, F. Del Ponte Hospital, Varese, Italy
| | - Yvan Vandenplas
- KidZ Health Castle, Free University of Brussels, Brussels, Belgium
| | - Gianvincenzo Zuccotti
- Department of Pediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy
| |
Collapse
|
21
|
Voigt J, Lele M. Lactobacillus rhamnosus Used in the Perinatal Period for the Prevention of Atopic Dermatitis in Infants: A Systematic Review and Meta-Analysis of Randomized Trials. Am J Clin Dermatol 2022; 23:801-811. [PMID: 36161401 PMCID: PMC9576646 DOI: 10.1007/s40257-022-00723-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2022] [Indexed: 12/03/2022]
Abstract
Background Numerous systematic reviews and meta-analyses have examined the effects of probiotics used perinatally on prevention or treatment of atopic disease in infants and children. However, to date, no review has examined randomized controlled trials of Lactobacillus rhamnosus, specifically, administered both prenatally and postnatally and its effect over a long period of time. Objective The objective was to determine if L. rhamnosus either used solely or in conjunction with other probiotics demonstrates a long-term preventive effect on atopic disease in pediatric patients when used perinatally. Methods A systematic review was undertaken to identify those studies where L. rhamnosus was used (either solely or in conjunction with other probiotics). The following databases were searched from the year 2000 through December 8, 2021: PubMed, Cochrane Reviews and Cochrane Central Database of Controlled Trials; systematic reviews were hand searched to identify randomized controlled trials (RCTs). Meta-analytic statistical techniques were then employed. Evaluation of the incidence of atopic eczema was also examined longitudinally based on timeframe. Grading of Recommendations, Assessment, Development and Evaluations (GRADE) assessments were employed to determine the quality of the evidence. Results Eleven randomized controlled trials were identified which examined L. rhamnosus in its effect on atopy. Risk of bias was low on the majority of the domains assessed. Meta-analysis of the timeframes ≤ 2 years (RR 0.60, 95% CI 0.47–0.75; p < 0.00001) and 6–7 years (RR 0.62, 95% CI 0.50–0.75; p < 0.00001) demonstrated statistically significant reductions in atopic eczema with use of L. rhamnosus. For the 4 to 5-year (RR 0.74, 95% CI 0.55–1.00; p = 0.05) and 10–11-year (RR 0.68, 95% CI 0.37–1.27; p = 0.23) timeframes there was no statistically significant reduction. GRADE assessment for each timeframe was considered moderate in two, owing to high attrition rates in all of the studies, and low in two due to imprecision. Conclusion Based on the meta-analysis and GRADE assessments, the use of L. rhamnosus with or without other probiotics appears to have a positive effect in reducing the incidence of atopic eczema in pediatric patients at least out to 7 years. Attrition rates temper these findings. Supplementary Information The online version contains supplementary material available at 10.1007/s40257-022-00723-x.
Collapse
Affiliation(s)
- Jeffrey Voigt
- Medical Device Consultants Ridgewood, LLC, 41 West Prospect St., Waldwick, NJ, 07463, USA.
| | - Meenal Lele
- Lil Mixins, 1711 North Howard St., Philadelphia, PA, 19122, USA
| |
Collapse
|
22
|
Saeki H, Ohya Y, Furuta J, Arakawa H, Ichiyama S, Katsunuma T, Katoh N, Tanaka A, Tsunemi Y, Nakahara T, Nagao M, Narita M, Hide M, Fujisawa T, Futamura M, Masuda K, Matsubara T, Murota H, Yamamoto-Hanada K. English Version of Clinical Practice Guidelines for the Management of Atopic Dermatitis 2021. J Dermatol 2022; 49:e315-e375. [PMID: 35996152 DOI: 10.1111/1346-8138.16527] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/03/2022] [Accepted: 06/17/2022] [Indexed: 11/29/2022]
Abstract
This is the English version of the Clinical Practice Guidelines for the Management of Atopic Dermatitis 2021. Atopic dermatitis (AD) is a disease characterized by relapsing eczema with pruritus as a primary lesion. In Japan, from the perspective of evidence-based medicine, the current strategies for the treatment of AD consist of three primary measures: (i) use of topical corticosteroids, tacrolimus ointment, and delgocitinib ointment as the main treatment of the inflammation; (ii) topical application of emollients to treat the cutaneous barrier dysfunction; and (iii) avoidance of apparent exacerbating factors, psychological counseling, and advice about daily life. In the present revised guidelines, descriptions of three new drugs, namely, dupilumab, delgocitinib, and baricitinib, have been added. The guidelines present recommendations to review clinical research articles, evaluate the balance between the advantages and disadvantages of medical activities, and optimize medical activity-related patient outcomes with respect to several important points requiring decision-making in clinical practice.
Collapse
Affiliation(s)
- Hidehisa Saeki
- Department of Dermatology, Nippon Medical School, Tokyo, Japan
| | - Yukihiro Ohya
- Allergy Center, National Center for Child Health and Development, Tokyo, Japan
| | - Junichi Furuta
- Medical Informatics and Management, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Hirokazu Arakawa
- Kitakanto Allergy Research Institute, Kibounoie Hospital, Midori, Japan
| | - Susumu Ichiyama
- Department of Dermatology, Nippon Medical School, Tokyo, Japan
| | - Toshio Katsunuma
- Department of Pediatrics, The Jikei University Daisan Hospital, Tokyo, Japan
| | - Norito Katoh
- Department of Dermatology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Akio Tanaka
- Department of Dermatology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yuichiro Tsunemi
- Department of Dermatology, Saitama Medical University, Saitama, Japan
| | - Takeshi Nakahara
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Mizuho Nagao
- Allergy Center, National Hospital Organization Mie National Hospital, Tsu, Japan
| | - Masami Narita
- Department of Pediatrics, Faculty of Medicine, Kyorin University, Tokyo, Japan
| | - Michihiro Hide
- Department of Dermatology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.,Department of Dermatology, Hiroshima Citizens Hospital, Hiroshima, Japan
| | - Takao Fujisawa
- Allergy Center, National Hospital Organization Mie National Hospital, Tsu, Japan
| | - Masaki Futamura
- Division of Pediatrics, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Koji Masuda
- Department of Dermatology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Tomoyo Matsubara
- Department of Pediatrics, Dokkyo Medical University Saitama Medical Center, Saitama, Japan
| | - Hiroyuki Murota
- Department of Dermatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | | |
Collapse
|
23
|
Hu Q, Acharya A, Leung WK, Pelekos G. Sponsorship Bias in Clinical Trials in the Dental Application of Probiotics: A Meta-Epidemiological Study. Nutrients 2022; 14:nu14163409. [PMID: 36014917 PMCID: PMC9413900 DOI: 10.3390/nu14163409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/06/2022] [Accepted: 08/10/2022] [Indexed: 11/16/2022] Open
Abstract
Many experimental and clinical trials have investigated the dental application of probiotics, although the evidence concerning the effects of probiotic supplements is conflicting. We aimed to examine whether sponsorship in trials about dental applications of probiotics is associated with biased estimates of treatment effects. Overall, 13 meta-analyses involving 48 randomized controlled trials (23 with high risk of sponsorship bias, 25 with low risk) with continuous outcomes were included. Effect sizes were calculated from differences in means of first reported continuous outcomes, divided by the pooled standard deviation. For each meta-analysis, the difference in standardized mean differences between high-risk and low-risk trials was estimated by random effects meta-regression. Differences in standardized mean differences (DSMDs) were then calculated via meta-analyses in a random effects meta-analysis model. A combined DSMD of greater than zero indicated that high-risk trials showed more significant treatment effects than low-risk trials. The results show that trials with a high risk of sponsorship bias showed more significant intervention effects than did low-risk trials (combined DSMD, 0.06; 95% confidence interval, 0.3 to 0.9; p < 0.001), with low heterogeneity among meta-analyses (I2 = 0%; between-meta-analyses variance τ2 = 0.00). Based on our study, high-risk clinical trials with continuous outcomes reported more favorable intervention effects than did low-risk trials in general.
Collapse
Affiliation(s)
- Qin Hu
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Aneesha Acharya
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR 999077, China
- Dr D. Y. Patil Dental College and Hospital, Dr D. Y. Patil Vidyapeeth, Pune 411018, India
| | - Wai Keung Leung
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR 999077, China
| | - George Pelekos
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR 999077, China
- Correspondence:
| |
Collapse
|
24
|
Uwaezuoke SN, Ayuk AC, Eze JN, Odimegwu CL, Ndiokwelu CO, Eze IC. Postnatal probiotic supplementation can prevent and optimize treatment of childhood asthma and atopic disorders: A systematic review of randomized controlled trials. Front Pediatr 2022; 10:956141. [PMID: 36061384 PMCID: PMC9437454 DOI: 10.3389/fped.2022.956141] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 08/04/2022] [Indexed: 11/13/2022] Open
Abstract
Background Although several randomized controlled trials (RCTs) published over the past 5 years show that prenatal or postnatal probiotics may prevent or optimize the treatment of childhood asthma and atopic disorders, findings from the systematic reviews and meta-analyses of these studies appear inconsistent. More recent RCTs have focused on postnatal probiotics, and linked specific probiotic strains to better disease outcomes. Objective This systematic review aimed to determine if postnatal probiotics are as effective as prenatal probiotics in preventing or treating childhood asthma and atopic disorders. Methods We searched the PubMed, Medline, Google Scholar, and EMBASE databases for RCTs published within the past 5 years (from 2017 to 2022). We included only full-text RCTs on human subjects published in or translated into the English language. We retrieved relevant data items with a preconceived data-extraction form and assessed the methodological quality of the selected RCTs using the Cochrane Collaboration's tool for assessing the risk of bias in randomized trials. We qualitatively synthesized the retrieved data to determine any significant differences in study endpoints of the probiotic and placebo groups. Results A total of 1,320 participants (688 and 632 in the probiotic and placebo groups) from six RCTs were investigated. One RCT showed that early Lactobacillus rhamnosus GG (LGG) led to a reduction in the cumulative incidence rate of asthma. Another study demonstrated that mixed strains of Lactobacillus paracasei and Lactobacillus fermentum could support clinical improvement in children with asthma while one trial reported a significant reduction in the frequency of asthma exacerbations using a mixture of Ligilactobacillus salivarius and Bifidobacterium breve. Three trials showed that a combination of LGG and Bifidobacterium animalis subsp lactis, Lactobacillus rhamnosus alone, and a probiotic mixture of Lactobacillus ŁOCK strains improved clinical outcomes in children with atopic dermatitis and cow-milk protein allergy. Conclusions Postnatal strain-specific probiotics (in single or mixed forms) are beneficial in preventing and treating atopic dermatitis and other allergies. Similarly, specific strains are more effective in preventing asthma or improving asthma outcomes. We recommend more interventional studies to establish the most useful probiotic strain in these allergic diseases.
Collapse
Affiliation(s)
- Samuel N. Uwaezuoke
- Department of Paediatrics, University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu, Nigeria
- Department of Pediatrics, College of Medicine, University of Nigeria, Ituku-Ozalla Enugu Campus, Enugu, Nigeria
| | - Adaeze C. Ayuk
- Department of Paediatrics, University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu, Nigeria
- Department of Pediatrics, College of Medicine, University of Nigeria, Ituku-Ozalla Enugu Campus, Enugu, Nigeria
| | - Joy N. Eze
- Department of Paediatrics, University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu, Nigeria
- Department of Pediatrics, College of Medicine, University of Nigeria, Ituku-Ozalla Enugu Campus, Enugu, Nigeria
| | - Chioma L. Odimegwu
- Department of Paediatrics, University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu, Nigeria
- Department of Pediatrics, College of Medicine, University of Nigeria, Ituku-Ozalla Enugu Campus, Enugu, Nigeria
| | - Chibuzo O. Ndiokwelu
- Department of Paediatrics, University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu, Nigeria
| | - Ikenna C. Eze
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| |
Collapse
|
25
|
Varela-Trinidad GU, Domínguez-Díaz C, Solórzano-Castanedo K, Íñiguez-Gutiérrez L, Hernández-Flores TDJ, Fafutis-Morris M. Probiotics: Protecting Our Health from the Gut. Microorganisms 2022; 10:1428. [PMID: 35889147 PMCID: PMC9316266 DOI: 10.3390/microorganisms10071428] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 02/07/2023] Open
Abstract
The gut microbiota (GM) comprises billions of microorganisms in the human gastrointestinal tract. This microbial community exerts numerous physiological functions. Prominent among these functions is the effect on host immunity through the uptake of nutrients that strengthen intestinal cells and cells involved in the immune response. The physiological functions of the GM are not limited to the gut, but bidirectional interactions between the gut microbiota and various extraintestinal organs have been identified. These interactions have been termed interorganic axes by several authors, among which the gut-brain, gut-skin, gut-lung, gut-heart, and gut-metabolism axes stand out. It has been shown that an organism is healthy or in homeostasis when the GM is in balance. However, altered GM or dysbiosis represents a critical factor in the pathogenesis of many local and systemic diseases. Therefore, probiotics intervene in this context, which, according to various published studies, allows balance to be maintained in the GM, leading to an individual's good health.
Collapse
Affiliation(s)
- Gael Urait Varela-Trinidad
- Doctorado en Ciencias Biomédicas, Con Orientaciones en Inmunología y Neurociencias, Universidad de Guadalajara, Sierra Mojada 950, Guadalajara 44340, Mexico; (G.U.V.-T.); (C.D.-D.)
- Centro de Investigación en Inmunología y Dermatología (CIINDE), Calzada del Federalismo Nte 3102, Zapopan 45190, Mexico
| | - Carolina Domínguez-Díaz
- Doctorado en Ciencias Biomédicas, Con Orientaciones en Inmunología y Neurociencias, Universidad de Guadalajara, Sierra Mojada 950, Guadalajara 44340, Mexico; (G.U.V.-T.); (C.D.-D.)
- Centro de Investigación en Inmunología y Dermatología (CIINDE), Calzada del Federalismo Nte 3102, Zapopan 45190, Mexico
| | - Karla Solórzano-Castanedo
- Doctorado en Ciencias de la Nutrición Traslacional, Universidad de Guadalajara, Sierra Mojada 950, Guadalajara 44340, Mexico;
| | - Liliana Íñiguez-Gutiérrez
- Instituto de Investigación de Inmunodeficiencias y VIH, Hospital Civil de Guadalajara, Coronel Calderón 777, Guadalajara 44280, Mexico; (L.Í.-G.); (T.d.J.H.-F.)
| | - Teresita de Jesús Hernández-Flores
- Instituto de Investigación de Inmunodeficiencias y VIH, Hospital Civil de Guadalajara, Coronel Calderón 777, Guadalajara 44280, Mexico; (L.Í.-G.); (T.d.J.H.-F.)
- Departamento de Disciplinas Filosóficas Metodológicas e Intrumentales, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Guadalajara 44340, Mexico
| | - Mary Fafutis-Morris
- Centro de Investigación en Inmunología y Dermatología (CIINDE), Calzada del Federalismo Nte 3102, Zapopan 45190, Mexico
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Guadalajara 44340, Mexico
| |
Collapse
|
26
|
Chen N, Liu F, Gao Q, Wang R, Zhang L, Li Y. A Meta-Analysis of Probiotics for the Treatment of Allergic Airway Diseases in Children and Adolescents. Am J Rhinol Allergy 2022; 36:480-490. [PMID: 35238209 DOI: 10.1177/19458924221080159] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Allergic airway disease is a chronic airway allergic inflammatory disease including allergic rhinitis (AR) and allergic asthma which is common in children and adolescents. Recently the probiotics has been becoming a supplementary or alternative therapy to allergic diseases, however the effect of them has not been clearly established. OBJECTIVE The purpose of the present meta-analysis was to evaluate the effectiveness of probiotics on allergic airway disease including AR and allergic asthma in children and adolescents. METHODS We performed a comprehensive search on PubMed, Cochrane Library, EMBASE for relevant publications from 1 Jan 2000 to 1 July 2021. Physical examinations, Pediatric Rhinoconjunctivitis Quality of Life Questionnaires (PRQLQs), Total Nasal Symptom Score (TNSS), Nasal or Eye Symptom Score (NSS or ESS), serum allergen-specific IgE, and eosinophil were used as evaluating indicators for AR and allergic asthma in children and adolescents. The meta-analysis was performed using Review Manager (RevMan, Version 5.3). RESULTS 15 randomized controlled trials (RCTs) with a total of 1388 participants were included for the meta-analysis. Among them, 729 patients treated with probiotics served as the probiotics group, and 659 patients with placebo as control group. Significantly greater reduction in PRQLQs from baseline to endpoint (SMD = -2.57, 95% CI [ - 4.66, -0.48] P < 0.01), NSS (SMD = -1.43, 95% CI [ - 1.63, - 1.23], P < 0.01) and ESS (total MD = -1.67, 95% CI [ - 1.79, - 1.55], P < 0.01) were observed in probiotics group compared to control group. Probiotics have no significant effect to serum IgE and eosinophils (P > 0.01). CONCLUSION The results of this meta-analysis indicated that probiotics treatment may reduce PRQLQs, NSS, ESS in patients with allergic airway disease. More research involving the mechanism of probiotics are needed to clarify the role of probiotics in AR and allergic asthma in children and adolescents.
Collapse
Affiliation(s)
- Na Chen
- Department of Otolaryngology, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Fang Liu
- Department of Otolaryngology, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Qiang Gao
- Department of Gastroenterology and Hepatology, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Rui Wang
- Department of Otolaryngology, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Lei Zhang
- Department of Otolaryngology, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Youwei Li
- Department of Radiology, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
27
|
Augustine T, Kumar M, Al Khodor S, van Panhuys N. Microbial Dysbiosis Tunes the Immune Response Towards Allergic Disease Outcomes. Clin Rev Allergy Immunol 2022:10.1007/s12016-022-08939-9. [PMID: 35648372 DOI: 10.1007/s12016-022-08939-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2022] [Indexed: 02/07/2023]
Abstract
The hygiene hypothesis has been popularized as an explanation for the rapid increase in allergic disease observed over the past 50 years. Subsequent epidemiological studies have described the protective effects that in utero and early life exposures to an environment high in microbial diversity have in conferring protective benefits against the development of allergic diseases. The rapid advancement in next generation sequencing technology has allowed for analysis of the diverse nature of microbial communities present in the barrier organs and a determination of their role in the induction of allergic disease. Here, we discuss the recent literature describing how colonization of barrier organs during early life by the microbiota influences the development of the adaptive immune system. In parallel, mechanistic studies have delivered insight into the pathogenesis of disease, by demonstrating the comparative effects of protective T regulatory (Treg) cells, with inflammatory T helper 2 (Th2) cells in the development of immune tolerance or induction of an allergic response. More recently, a significant advancement in our understanding into how interactions between the adaptive immune system and microbially derived factors play a central role in the development of allergic disease has emerged. Providing a deeper understanding of the symbiotic relationship between our microbiome and immune system, which explains key observations made by the hygiene hypothesis. By studying how perturbations that drive dysbiosis of the microbiome can cause allergic disease, we stand to benefit by delineating the protective versus pathogenic aspects of human interactions with our microbial companions, allowing us to better harness the use of microbial agents in the design of novel prophylactic and therapeutic strategies.
Collapse
Affiliation(s)
- Tracy Augustine
- Laboratory of Immunoregulation, Sidra Medicine, PO BOX 26999, Doha, Qatar
| | - Manoj Kumar
- Microbiome and Host-Microbes Interactions Laboratory, Sidra Medicine, Doha, Qatar
| | - Souhaila Al Khodor
- Microbiome and Host-Microbes Interactions Laboratory, Sidra Medicine, Doha, Qatar
| | | |
Collapse
|
28
|
Anania C, Brindisi G, Martinelli I, Bonucci E, D’Orsi M, Ialongo S, Nyffenegger A, Raso T, Spatuzzo M, De Castro G, Zicari AM, Carraro C, Piccioni MG, Olivero F. Probiotics Function in Preventing Atopic Dermatitis in Children. Int J Mol Sci 2022; 23:ijms23105409. [PMID: 35628229 PMCID: PMC9141149 DOI: 10.3390/ijms23105409] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/04/2022] [Accepted: 05/07/2022] [Indexed: 11/16/2022] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disorder characterized by relapsing eczematous injuries and severe pruritus. In the last few years, the AD prevalence has been increasing, reaching 20% in children and 10% in adults in high-income countries. Recently, the potential role of probiotics in AD prevention has generated considerable interest. As many clinical studies show, the gut microbiota is able to modulate systemic inflammatory and immune responses influencing the development of sensitization and allergy. Probiotics are used increasingly against AD. However, the molecular mechanisms underlying the probiotics mediated anti-allergic effect remain unclear and there is controversy about their efficacy. In this narrative review, we examine the actual evidence on the effect of probiotic supplementation for AD prevention in the pediatric population, discussing also the potential biological mechanisms of action in this regard.
Collapse
Affiliation(s)
- Caterina Anania
- Department of Mother-Child, Urological Science, Sapienza University of Rome, 00161 Rome, Italy; (G.B.); (I.M.); (E.B.); (M.D.); (S.I.); (A.N.); (T.R.); (M.S.); (G.D.C.); (A.M.Z.); (C.C.); (M.G.P.)
- Correspondence:
| | - Giulia Brindisi
- Department of Mother-Child, Urological Science, Sapienza University of Rome, 00161 Rome, Italy; (G.B.); (I.M.); (E.B.); (M.D.); (S.I.); (A.N.); (T.R.); (M.S.); (G.D.C.); (A.M.Z.); (C.C.); (M.G.P.)
| | - Ivana Martinelli
- Department of Mother-Child, Urological Science, Sapienza University of Rome, 00161 Rome, Italy; (G.B.); (I.M.); (E.B.); (M.D.); (S.I.); (A.N.); (T.R.); (M.S.); (G.D.C.); (A.M.Z.); (C.C.); (M.G.P.)
| | - Edoardo Bonucci
- Department of Mother-Child, Urological Science, Sapienza University of Rome, 00161 Rome, Italy; (G.B.); (I.M.); (E.B.); (M.D.); (S.I.); (A.N.); (T.R.); (M.S.); (G.D.C.); (A.M.Z.); (C.C.); (M.G.P.)
| | - Miriam D’Orsi
- Department of Mother-Child, Urological Science, Sapienza University of Rome, 00161 Rome, Italy; (G.B.); (I.M.); (E.B.); (M.D.); (S.I.); (A.N.); (T.R.); (M.S.); (G.D.C.); (A.M.Z.); (C.C.); (M.G.P.)
| | - Sara Ialongo
- Department of Mother-Child, Urological Science, Sapienza University of Rome, 00161 Rome, Italy; (G.B.); (I.M.); (E.B.); (M.D.); (S.I.); (A.N.); (T.R.); (M.S.); (G.D.C.); (A.M.Z.); (C.C.); (M.G.P.)
| | - Anna Nyffenegger
- Department of Mother-Child, Urological Science, Sapienza University of Rome, 00161 Rome, Italy; (G.B.); (I.M.); (E.B.); (M.D.); (S.I.); (A.N.); (T.R.); (M.S.); (G.D.C.); (A.M.Z.); (C.C.); (M.G.P.)
| | - Tonia Raso
- Department of Mother-Child, Urological Science, Sapienza University of Rome, 00161 Rome, Italy; (G.B.); (I.M.); (E.B.); (M.D.); (S.I.); (A.N.); (T.R.); (M.S.); (G.D.C.); (A.M.Z.); (C.C.); (M.G.P.)
| | - Mattia Spatuzzo
- Department of Mother-Child, Urological Science, Sapienza University of Rome, 00161 Rome, Italy; (G.B.); (I.M.); (E.B.); (M.D.); (S.I.); (A.N.); (T.R.); (M.S.); (G.D.C.); (A.M.Z.); (C.C.); (M.G.P.)
| | - Giovanna De Castro
- Department of Mother-Child, Urological Science, Sapienza University of Rome, 00161 Rome, Italy; (G.B.); (I.M.); (E.B.); (M.D.); (S.I.); (A.N.); (T.R.); (M.S.); (G.D.C.); (A.M.Z.); (C.C.); (M.G.P.)
| | - Anna Maria Zicari
- Department of Mother-Child, Urological Science, Sapienza University of Rome, 00161 Rome, Italy; (G.B.); (I.M.); (E.B.); (M.D.); (S.I.); (A.N.); (T.R.); (M.S.); (G.D.C.); (A.M.Z.); (C.C.); (M.G.P.)
| | - Carlo Carraro
- Department of Mother-Child, Urological Science, Sapienza University of Rome, 00161 Rome, Italy; (G.B.); (I.M.); (E.B.); (M.D.); (S.I.); (A.N.); (T.R.); (M.S.); (G.D.C.); (A.M.Z.); (C.C.); (M.G.P.)
| | - Maria Grazia Piccioni
- Department of Mother-Child, Urological Science, Sapienza University of Rome, 00161 Rome, Italy; (G.B.); (I.M.); (E.B.); (M.D.); (S.I.); (A.N.); (T.R.); (M.S.); (G.D.C.); (A.M.Z.); (C.C.); (M.G.P.)
| | - Francesca Olivero
- Pediatric Clinic, Department of Pediatrics, Fondazione IRCSS Policlinico San Matteo, University of Pavia, 27100 Pavia, Italy;
| |
Collapse
|
29
|
Colquitt AS, Miles EA, Calder PC. Do Probiotics in Pregnancy Reduce Allergies and Asthma in Infancy and Childhood? A Systematic Review. Nutrients 2022; 14:nu14091852. [PMID: 35565819 PMCID: PMC9105059 DOI: 10.3390/nu14091852] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 02/01/2023] Open
Abstract
The maternal immune system is very important in the development of the foetal immune system. Probiotics have been shown to help regulate immune responses. Therefore, it is possible that the administration of probiotics to pregnant women could influence the development of the foetal immune system, reducing the likelihood of infants and children developing an allergic condition. The aim of this research was to conduct a systematic review to determine whether administering probiotics to pregnant women can reduce the incidence of allergic disease in their children. Medline, CINAHL and Embase databases were searched for randomised controlled trials (RCTs) that compared supplementation of probiotics to pregnant women to a placebo control and recorded the presentation of allergic conditions in their children. Data extracted from the study reports included their characteristics and findings. Study quality and risk of bias were assessed. From a total of 850 articles identified in the search, 6 were eligible for inclusion in this review. Two studies found no effect of maternal probiotics on the outcomes measured, two studies found that the incidence of eczema or atopic dermatitis (AD) was reduced by maternal probiotics, one study found no effect on the overall incidence of atopic sensitisation, but a reduction in a subgroup of children at high hereditary risk of allergic disease, and one study found no effect in an intention to treat analysis, but a reduction in AD in complete case analysis. The results of these studies are inconsistent but demonstrate that probiotics may have the potential to reduce infant allergies when administered prenatally, particularly in children at high risk of allergy development. There is a need for further larger-scale studies to be performed in order to provide a more definitive answer. Such studies should focus on at-risk groups.
Collapse
Affiliation(s)
- Alexander S. Colquitt
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (A.S.C.); (E.A.M.)
| | - Elizabeth A. Miles
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (A.S.C.); (E.A.M.)
| | - Philip C. Calder
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (A.S.C.); (E.A.M.)
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton SO16 6YD, UK
- Correspondence:
| |
Collapse
|
30
|
Feng T, Liu Y. Microorganisms in the reproductive system and probiotic's regulatory effects on reproductive health. Comput Struct Biotechnol J 2022; 20:1541-1553. [PMID: 35465162 PMCID: PMC9010680 DOI: 10.1016/j.csbj.2022.03.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 12/18/2022] Open
Abstract
The presence of microbial communities in the reproductive tract has been revealed, and this resident microbiota is involved in the maintenance of health. Intentional modulation via probiotics has been proposed as a possible strategy to enhance reproductive health and reduce the risk of diseases. The male seminal microbiota has been suggested as an important factor that influences a couple’s health, pregnancy outcomes, and offspring health. Probiotics have been reported to play a role in male fertility and to affect the health of mothers and offspring. While the female reproductive microbiota is more complicated and has been identified in both the upper and lower reproductive systems, they together contribute to health maintenance. Probiotics have shown regulatory effects on the female reproductive tract, thereby contributing to homeostasis of the tract and influencing the health of offspring. Further, through transmission of bacteria or through other indirect mechanisms, the parent’s reproductive microbiota and probiotic intervention influence infant gut colonization and immunity development, with potential health consequences. In vitro and in vivo studies have explored the mechanisms underlying the benefits of probiotic administration and intervention, and an array of positive results, such as modulation of microbiota composition, regulation of metabolism, promotion of the epithelial barrier, and improvement of immune function, have been observed. Herein, we review the state of the art in reproductive system microbiota and its role in health and reproduction, as well as the beneficial effects of probiotics on reproductive health and their contributions to the prevention of associated diseases.
Collapse
|
31
|
Hui-Beckman J, Kim BE, Leung DY. Origin of Allergy From In Utero Exposures to the Postnatal Environment. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2022; 14:8-20. [PMID: 34983104 PMCID: PMC8724834 DOI: 10.4168/aair.2022.14.1.8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/28/2021] [Accepted: 07/11/2021] [Indexed: 04/28/2023]
Abstract
As the incidence of atopic conditions continues to increase, emphasis has been placed on understanding the origin of allergy with hope that prevention measures can be achieved. The perinatal environment is important for this understanding, given that both the immune system and microbiome start forming prenatally. Maternal exposure can greatly impact on fetal health. Additionally, the dysfunctional epithelial barrier is influential in allowing allergens and irritants to penetrate the skin or mucosa, leading to the release of proinflammatory cytokines and mediators to drive type 2 tissue inflammation and the onset of allergy. There are numerous factors related to skin, airway, and gut epithelial barriers dysfunction, and genetic predispositions are also present. Comprehensive birth cohort studies and further mechanistic studies will be keys to understanding the origin of allergy.
Collapse
Affiliation(s)
| | - Byung Eui Kim
- Department of Pediatrics, National Jewish Health, Denver, CO 80206, USA
| | - Donald Ym Leung
- Department of Pediatrics, National Jewish Health, Denver, CO 80206, USA.
| |
Collapse
|
32
|
Turner RB, Lehtoranta L, Hibberd A, Männikkö S, Zabel B, Yeung N, Huttunen T, Burns FR, Lehtinen MJ. Effect of Bifidobacterium animalis spp. lactis Bl-04 on Rhinovirus-Induced Colds: A Randomized, Placebo-Controlled, Single-Center, Phase II Trial in Healthy Volunteers. EClinicalMedicine 2022; 43:101224. [PMID: 34927036 PMCID: PMC8649651 DOI: 10.1016/j.eclinm.2021.101224] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 11/11/2021] [Accepted: 11/17/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND This study was designed to assess the efficacy of Bifidobacterium animalis ssp. lactis (Bl-04) for prevention of rhinovirus colds and to explore the interactions between the probiotic, the viral infection, the host response and the host microbiome. METHODS The effect of ingestion of the probiotic Bl-04 was evaluated in a randomized, double-blinded rhinovirus (RV) challenge study. Healthy volunteers recruited from a university community in USA were randomized 1:1 using a computer generated code to ingest either Bl-04 (n=165) or placebo (n=169) for 28 days and were then challenged with RV-A39, and followed for 14 days. All study interactions and sample collection occurred in dedicated clinical research space. The primary analysis was the effect of the probiotic on the incidence of RV-associated illness. (Trial registration: NCT02679807, study complete). FINDINGS The first cohort of volunteers was randomized on March 14, 2016 and the last (5th) cohort was randomized on March 12, 2018. Sixty-three (56%, 95% CI [47%; 66%]) of the 112 subjects in the active group and 60 (50%,95% CI [41%; 59%]) of the 120 subjects in the placebo group had a protocol-defined rhinovirus-associated illness (χ2=0·91, p=0·34). The point estimate of the difference in illness (active-placebo) is 6.3% (95% CI -6.7;19.1). There were no adverse events that were judged as definitely or probably related to the study product. INTERPRETATION In this study there was no effect of orally administered Bl-04 on the occurrence of RV-associated illness. FUNDING Danisco Sweeteners Oy (now IFF Health & Biosciences).
Collapse
Affiliation(s)
- Ronald B. Turner
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA
- Corresponding To: 512 Rosemont Drive, Charlottesville, VA 22903
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Ashraf SA, Elkhalifa AEO, Ahmad MF, Patel M, Adnan M, Sulieman AME. Probiotic Fermented Foods and Health Promotion. AFRICAN FERMENTED FOOD PRODUCTS- NEW TRENDS 2022:59-88. [DOI: 10.1007/978-3-030-82902-5_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
34
|
Cuinat C, Stinson SE, Ward WE, Comelli EM. Maternal Intake of Probiotics to Program Offspring Health. Curr Nutr Rep 2022; 11:537-562. [PMID: 35986890 PMCID: PMC9750916 DOI: 10.1007/s13668-022-00429-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2022] [Indexed: 01/31/2023]
Abstract
PURPOSE OF REVIEW Probiotics intake may be considered beneficial by prospective and pregnant mothers, but their effects on offspring development are incompletely understood. The purpose of this review was to examine recent pre-clinical and clinical studies to understand how maternal probiotics exposure affects offspring health outcomes. RECENT FINDINGS Effects were investigated in the context of supporting offspring growth, intestinal health, and gut microbiota, preventing allergic diseases, supporting neurodevelopment, and preventing metabolic disorders in pre-clinical and clinical studies. Most human studies focused on infancy outcomes, whereas pre-clinical studies also examined outcomes at adolescence and young adulthood. While still understudied, both pre-clinical and clinical studies propose epigenetic modifications as an underlying mechanism. Optimal timing of intervention remains unclear. Administration of selected probiotics to mothers has programming potential for sustaining life-long health of offspring. Administration protocols, specific windows of susceptibility, and individual-specific responses need to be further studied.
Collapse
Affiliation(s)
- Céline Cuinat
- grid.17063.330000 0001 2157 2938Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | - Sara E. Stinson
- grid.17063.330000 0001 2157 2938Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | - Wendy E. Ward
- grid.17063.330000 0001 2157 2938Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON Canada ,grid.411793.90000 0004 1936 9318Department of Kinesiology, Faculty of Applied Health Sciences, Brock University, St. Catharines, ON Canada
| | - Elena M. Comelli
- grid.17063.330000 0001 2157 2938Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON Canada ,grid.411793.90000 0004 1936 9318Department of Kinesiology, Faculty of Applied Health Sciences, Brock University, St. Catharines, ON Canada ,grid.17063.330000 0001 2157 2938Joannah and Brian Lawson Centre for Child Nutrition, Faculty of Medicine, University of Toronto, Toronto, ON Canada
| |
Collapse
|
35
|
Current Insights into Atopic March. CHILDREN (BASEL, SWITZERLAND) 2021; 8:children8111067. [PMID: 34828780 PMCID: PMC8620020 DOI: 10.3390/children8111067] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/10/2021] [Accepted: 11/16/2021] [Indexed: 11/20/2022]
Abstract
The incidence of allergic diseases is increasing, and research on their epidemiology, pathophysiology, and the prevention of onset is urgently needed. The onset of allergic disease begins in infancy with atopic dermatitis and food allergy and develops into allergic asthma and allergic rhinitis in childhood; the process is defined as “atopic march”. Atopic march is caused by multiple immunological pathways, including allergen exposure, environmental pollutants, skin barrier dysfunction, type 2 inflammation, and oxidative stress, which promote the progression of atopic march. Using recent evidence, herein, we explain the involvement of allergic inflammatory conditions and oxidative stress in the process of atopic march, its epidemiology, and methods for prevention of onset.
Collapse
|
36
|
Hu T, Dong Y, Yang C, Zhao M, He Q. Pathogenesis of Children's Allergic Diseases: Refocusing the Role of the Gut Microbiota. Front Physiol 2021; 12:749544. [PMID: 34721073 PMCID: PMC8551706 DOI: 10.3389/fphys.2021.749544] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/24/2021] [Indexed: 12/26/2022] Open
Abstract
Allergic diseases comprise a genetically heterogeneous cluster of immunologically mediated diseases, including asthma, food allergy (FA), allergic rhinitis (AR) and eczema, that have become major worldwide health problems. Over the past few decades, the spread of allergic diseases has displayed an increasing trend, and it has been reported that 22% of 1.39 billion people in 30 countries have a type of allergic disease. Undoubtedly, allergic diseases, which can be chronic, with significant morbidity, mortality and dynamic progression, impose major economic burdens on society and families; thus, exploring the cause of allergic diseases and reducing their prevalence is a top priority. Recently, it has been reported that the gastrointestinal (GI) microbiota can provide vital signals for the development, function, and regulation of the immune system, and the above-mentioned contributions make the GI microbiota a key player in allergic diseases. Notably, the GI microbiota is highly influenced by the mode of delivery, infant diet, environment, antibiotic use and so on. Specifically, changes in the environment can result in the dysbiosis of the GI microbiota. The proper function of the GI microbiota depends on a stable cellular composition which in the case of the human microbiota consists mainly of bacteria. Large shifts in the ratio between these phyla or the expansion of new bacterial groups lead to a disease-promoting imbalance, which is often referred to as dysbiosis. And the dysbiosis can lead to alterations of the composition of the microbiota and subsequent changes in metabolism. Further, the GI microbiota can affect the physiological characteristics of the human host and modulate the immune response of the host. The objectives of this review are to evaluate the development of the GI microbiota, the main drivers of the colonization of the GI tract, and the potential role of the GI microbiota in allergic diseases and provide a theoretical basis as well as molecular strategies for clinical practice.
Collapse
Affiliation(s)
- Tingting Hu
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yinmiao Dong
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Chenghao Yang
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Mingyi Zhao
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Qingnan He
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
37
|
Tramper‐Stranders G, Ambrożej D, Arcolaci A, Atanaskovic‐Markovic M, Boccabella C, Bonini M, Karavelia A, Mingomataj E, O' Mahony L, Sokolowska M, Untersmayr E, Feleszko W. Dangerous liaisons: Bacteria, antimicrobial therapies, and allergic diseases. Allergy 2021; 76:3276-3291. [PMID: 34390006 DOI: 10.1111/all.15046] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 07/31/2021] [Indexed: 12/15/2022]
Abstract
Microbiota composition and associated metabolic activities are essential for the education and development of a healthy immune system. Microbial dysbiosis, caused by risk factors such as diet, birth mode, or early infant antimicrobial therapy, is associated with the inception of allergic diseases. In turn, allergic diseases increase the risk for irrational use of antimicrobial therapy. Microbial therapies, such as probiotics, have been studied in the prevention and treatment of allergic diseases, but evidence remains limited due to studies with high heterogeneity, strain-dependent effectiveness, and variable outcome measures. In this review, we sketch the relation of microbiota with allergic diseases, the overuse and rationale for the use of antimicrobial agents in allergic diseases, and current knowledge concerning the use of bacterial products in allergic diseases. We urgently recommend 1) limiting antibiotic therapy in pregnancy and early childhood as a method contributing to the reduction of the allergy epidemic in children and 2) restricting antibiotic therapy in exacerbations and chronic treatment of allergic diseases, mainly concerning asthma and atopic dermatitis. Future research should be aimed at antibiotic stewardship implementation strategies and biomarker-guided therapy, discerning those patients that might benefit from antibiotic therapy.
Collapse
Affiliation(s)
- Gerdien Tramper‐Stranders
- Department of Pediatrics Franciscus Gasthuis & Vlietland Rotterdam the Netherlands
- Department of Neonatology Erasmus Medical CenterSophia Children's Hospital Rotterdam the Netherlands
| | - Dominika Ambrożej
- Department of Pediatric Pneumonology and Allergy Medical University of Warsaw Warsaw Poland
- Doctoral School Medical University of Warsaw Warsaw Poland
| | - Alessandra Arcolaci
- Immunology Unit University of Verona and General Hospital Borgo Roma Hospital Verona Italy
| | | | - Cristina Boccabella
- Department of Cardiovascular and Thoracic Sciences Università Cattolica del Sacro CuoreFondazione Policlinico Universitario A. Gemelli – IRCCS Rome Italy
| | - Matteo Bonini
- Department of Cardiovascular and Thoracic Sciences Università Cattolica del Sacro CuoreFondazione Policlinico Universitario A. Gemelli – IRCCS Rome Italy
- National Heart and Lung Institute (NHLI) Imperial College London London UK
| | - Aspasia Karavelia
- Department of Ear‐Nose‐Throat surgery General Hospital of Kozani Kozani Greece
| | - Ervin Mingomataj
- Department of Allergology & Clinical Immunology ‘Mother Theresa’ School of Medicine Tirana Albania
| | - Liam O' Mahony
- Departments of Medicine and Microbiology APC Microbiome IrelandNational University of Ireland Cork Ireland
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Zurich Switzerland
| | - Eva Untersmayr
- Institute of Pathophysiology and Allergy Research Center for Pathophysiology, Infectiology and Immunology Medical University of Vienna Vienna Austria
| | - Wojciech Feleszko
- Department of Pediatric Pneumonology and Allergy Medical University of Warsaw Warsaw Poland
| | | |
Collapse
|
38
|
Ayasse MT, Ahmed A, Espinosa ML, Walker CJ, Yousaf M, Thyssen JP, Silverberg JI. What are the highest yielding search strategy terms for systematic reviews in atopic dermatitis? A systematic review. Arch Dermatol Res 2021; 313:737-750. [PMID: 33221950 DOI: 10.1007/s00403-020-02165-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/16/2020] [Accepted: 10/30/2020] [Indexed: 12/30/2022]
Abstract
The impact of search strategies on systematic reviews (SR) of atopic dermatitis (AD) is unknown. The purpose of this review was to evaluate search strategies used in SR of AD and their impact on the frequency of manuscripts identified. MEDLINE and EMBASE were searched for SR related to AD. Simulations were performed by running combinations of search terms in MEDLINE and EMBASE. Overall, 250 SR met inclusion criteria, of which 225 specified search strategies. SR using 5-6 terms (20.0% to 12.1%) or ≥ 7 (40.0% to 18.8%) terms decreased, whereas SR using 3-4 terms numerically increased (18.8% to 30.2%) and 1-2 terms remained similar (37.5% to 38.9%) from 1999-2009 to 2015-2019. The most commonly searched terms were "atopic dermatitis" (n = 166), followed by "eczema" (n = 156), "dermatitis atopic'" (n = 81), "atopic eczema" (n = 74), "neurodermatitis" (n = 59), "Besniers prurigo" (n = 29), "infantile eczema" (n = 27), and "childhood eczema" (n = 19). Simulations revealed that "eczema" and "atopic dermatitis" yielded the most hits. The number of search terms that maximized hits in MEDLINE and EMBASE was 5 and 4, respectively. Search strategies for AD were heterogeneous, with high proportions of search strategies providing few search hits. Future studies should use standardized and optimized search terms.
Collapse
Affiliation(s)
- Marissa T Ayasse
- Department of Dermatology, The George Washington University School of Medicine and Health Sciences, 2150 Pennsylvania Avenue NW, Suite 2B-425, Washington, DC, 20037, USA
| | - Adnan Ahmed
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Maria L Espinosa
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Christina J Walker
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Muhammad Yousaf
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jacob P Thyssen
- Department of Dermatology and Allergy, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Jonathan I Silverberg
- Department of Dermatology, The George Washington University School of Medicine and Health Sciences, 2150 Pennsylvania Avenue NW, Suite 2B-425, Washington, DC, 20037, USA.
| |
Collapse
|
39
|
Abstract
The neonatal body provides a range of potential habitats, such as the gut, for microbes. These sites eventually harbor microbial communities (microbiotas). A "complete" (adult) gut microbiota is not acquired by the neonate immediately after birth. Rather, the exclusive, milk-based nutrition of the infant encourages the assemblage of a gut microbiota of low diversity, usually dominated by bifidobacterial species. The maternal fecal microbiota is an important source of bacterial species that colonize the gut of infants, at least in the short-term. However, development of the microbiota is influenced by the use of human milk (breast feeding), infant formula, preterm delivery of infants, caesarean delivery, antibiotic administration, family details and other environmental factors. Following the introduction of weaning (complementary) foods, the gut microbiota develops in complexity due to the availability of a diversity of plant glycans in fruits and vegetables. These glycans provide growth substrates for the bacterial families (such as members of the Ruminococcaceae and Lachnospiraceae) that, in due course, will dominate the gut microbiota of the adult. Although current data are often fragmentary and observational, it can be concluded that the nutrition that a child receives in early life is likely to impinge not only on the development of the microbiota at that time but also on the subsequent lifelong, functional relationships between the microbiota and the human host. The purpose of this review, therefore, is to discuss the importance of promoting the assemblage of functionally robust gut microbiotas at appropriate times in early life.
Collapse
Affiliation(s)
- Gerald W. Tannock
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
40
|
Childs CE, Munblit D, Ulfman L, Gómez-Gallego C, Lehtoranta L, Recker T, Salminen S, Tiemessen M, Collado MC. Potential Biomarkers, Risk Factors and their Associations with IgE-mediated Food Allergy in Early Life: A Narrative Review. Adv Nutr 2021; 13:S2161-8313(22)00081-3. [PMID: 34596662 PMCID: PMC8970818 DOI: 10.1093/advances/nmab122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Food allergy affects the quality of life of millions of people worldwide and presents a significant psychological and financial burden for both national and international public health. In the past few decades, the prevalence of allergic disease has been on the rise worldwide. Identified risk factors for food allergy include family history, mode of delivery, variations in infant feeding practices, prior diagnosis of other atopic diseases such as eczema, and social economic status. Identifying reliable biomarkers which predict the risk of developing food allergy in early life would be valuable in both preventing morbidity and mortality and by making current interventions available at the earliest opportunity. There is also the potential to identify new therapeutic targets. This narrative review provides details on the genetic, epigenetic, dietary and microbiome influences upon the development of food allergy and synthesizes the currently available data indicating potential biomarkers. While there is a large body of research evidence available within each field of potential risk factors, there are very limited number of studies which span multiple methodological fields, for example including immunology, microbiome, genetic/epigenetic factors and dietary assessment. We recommend that further collaborative research with detailed cohort phenotyping is required to identify biomarkers, and whether these vary between at-risk populations and the wider population. The low incidence of oral food challenge confirmed food allergy in the general population, and the complexities of designing nutritional intervention studies will provide challenges for researchers to address in generating high quality, reliable and reproducible research findings. STATEMENT OF SIGNIFICANCE Food allergy affects the quality of life of millions of people worldwide and presents a significant psychological and financial burden for both national and international public health. Identifying reliable biomarkers which predict the risk of developing food allergy would be valuable in both preventing morbidity and mortality and by making current interventions available at the earliest opportunity. This review provides details on the genetic, epigenetic, dietary and microbiome influences upon the development of food allergy. This helps in identifying reliable biomarkers to predict the risk of developing food allergy, which could be valuable in both preventing morbidity and mortality and by making interventions available at the earliest opportunity.
Collapse
Affiliation(s)
- Caroline E Childs
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom,Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Daniel Munblit
- Imperial College London, London, United Kingdom,Department of Paediatrics and Paediatric Infectious Diseases, Institute of Child’s Health, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia,Inflammation, Repair and Development Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | | | - Carlos Gómez-Gallego
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | | | | | | | | | | |
Collapse
|
41
|
Tan-Lim CSC, Esteban-Ipac NAR, Recto MST, Castor MAR, Casis-Hao RJ, Nano ALM. Comparative effectiveness of probiotic strains on the prevention of pediatric atopic dermatitis: A systematic review and network meta-analysis. Pediatr Allergy Immunol 2021; 32:1255-1270. [PMID: 33811784 DOI: 10.1111/pai.13514] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 02/23/2021] [Accepted: 03/30/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Atopic dermatitis is the most common chronic skin disease affecting the pediatric population. Probiotics have been proposed to be effective in preventing the development of pediatric atopic dermatitis. Although studies show promise for the use of probiotics, the evidence is still inconclusive due to significant heterogeneity and imprecision. OBJECTIVE To determine the comparative effectiveness of the different types of probiotic strains in preventing the development of atopic dermatitis among pediatric patients. METHODOLOGY A systematic search of Cochrane Library, MEDLINE, TRIP Database, and Centre for Research and Dissemination was conducted. Manual search of the reference lists and search for unpublished articles were also done. All randomized controlled trials available from inception until April 12, 2020, on the use of probiotics in the prevention of atopic dermatitis among children were included. The comparator groups considered are other probiotic strains and placebo. The primary outcome of interest was the development of atopic dermatitis. Two authors independently searched for articles, screened the articles for inclusion, appraised the articles using the Cochrane risk of bias tool version 2, and extracted the data. In case of disagreement, the two authors discussed the source of disagreement until consensus was reached. If consensus was not reached, an independent third party reviewer was consulted. Frequentist network meta-analysis was conducted using STATA 14 software. The ranking probabilities and surface under the cumulative ranking curve (SUCRA) values were obtained to determine ranking of the different probiotic strains based on efficacy and safety data. RESULTS We included 21 original studies represented by 35 records and a total of 5406 children with atopic dermatitis as diagnosed by clinicians or fulfillment of validated diagnostic criteria. All studies were randomized placebo-controlled trials. The top 3 probiotic preparations in terms of efficacy in reducing the risk of atopic dermatitis are Mix8 (Lactobacillus paracasei ST11, Bifidobacterium longum BL999), LP (Lactobacillus paracasei ssp paracasei F19) and Mix3 (Lactobacillus rhamnosus GG, Bifidobacterium animalis ssp lactis Bb-12). Mix8 compared with placebo probably reduces the risk of atopic dermatitis based on low-quality evidence (RR = 0.46, 95% CI 0.25-0.85). Mix3 compared with placebo also probably reduces the risk of atopic dermatitis based on low-quality evidence (RR = 0.50, 95% CI 0.27-0.94). It is uncertain whether LP compared with placebo reduces the risk of atopic dermatitis due to very-low-quality certainty of evidence (RR = 0.49, 95% CI 0.20-1.19). In terms of adverse events, LGG may slightly lead to less adverse events compared with placebo based on low-quality evidence (RR = 0.70, 95% CI 0.32-1.52). Mix4 may slightly lead to more adverse events compared with placebo based on low-quality evidence (RR = 1.06, 95% CI 0.02-51.88). Based on subgroup analysis of studies involving infants, Mix3 compared with placebo probably reduces the risk of atopic dermatitis based on low-quality evidence (RR = 0.46, 95% CI 0.22-0.97). In the subgroup analysis of studies where probiotics were administered to pregnant women and to infants, LRH compared with placebo probably reduces the risk of atopic dermatitis based on moderate-quality evidence (RR = 0.54, 95% CI 0.26-1.11). CONCLUSION Certain probiotic preparations demonstrate efficacy in reducing the risk of developing atopic dermatitis when administered to pregnant women, infants, or both.
Collapse
Affiliation(s)
- Carol Stephanie C Tan-Lim
- Department of Clinical Epidemiology, College of Medicine, University of the Philippines Manila, Manila, Philippines.,Department of Pediatrics, Division of Allergy and Immunology, University of the Philippines Manila - Philippine General Hospital, Manila, Philippines
| | - Natasha Ann R Esteban-Ipac
- Department of Clinical Epidemiology, College of Medicine, University of the Philippines Manila, Manila, Philippines
| | - Marysia Stella T Recto
- Department of Pediatrics, Division of Allergy and Immunology, University of the Philippines Manila - Philippine General Hospital, Manila, Philippines
| | - Mary Anne R Castor
- Department of Pediatrics, Division of Allergy and Immunology, University of the Philippines Manila - Philippine General Hospital, Manila, Philippines
| | - Roxanne J Casis-Hao
- Department of Pediatrics, Division of Allergy and Immunology, University of the Philippines Manila - Philippine General Hospital, Manila, Philippines
| | - Aimee Lou M Nano
- Department of Pediatrics, Division of Allergy and Immunology, University of the Philippines Manila - Philippine General Hospital, Manila, Philippines
| |
Collapse
|
42
|
Takahashi T, Fukudome H, Ueno HM, Watanabe-Matsuhashi S, Nakano T, Kobayashi T, Ishimaru K, Nakao A. Probiotic Supplementation and Human Milk Cytokine Profiles in Japanese Women: A Retrospective Study from an Open-Label Pilot Study. Nutrients 2021; 13:nu13072285. [PMID: 34209459 PMCID: PMC8308220 DOI: 10.3390/nu13072285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/24/2021] [Accepted: 06/28/2021] [Indexed: 11/26/2022] Open
Abstract
The benefits of probiotic supplementation to lactating mothers on human milk cytokines are inconclusive. Thus, we performed a comprehensive open-label pilot trial analysis of 27 human milk cytokines in lactating women with allergies (one to three months postpartum) to determine the effect of supplementation with a mixture of new probiotic strains. Participants voluntarily joined the probiotic (n = 41) or no supplementation control (n = 19) groups. The probiotic group took three probiotic tablets (Lactobacillus casei LC5, Bifidobacterium longum BG7, and Bacillus coagulans SANK70258) daily for one to three months postpartum. Milk samples were collected at one, two, and three months postpartum, and cytokine levels were measured using multiplex assays. The effects were analyzed using multivariate regression models. Eleven cytokines showed a positive rate of over 50% in the milk samples throughout testing in both groups. The positive rates of IL-1 receptor antagonist and IL-7 changed significantly with lactation progression in logistic regression models after adjusting for time and supplementation, whereas rates of other cytokines showed no significant differences. The lactational change patterns of IL-10 concentrations differed significantly between the two groups. A short-term supplementation of probiotics affects human milk cytokine levels in lactating women with a possible placebo effect still existing. Future placebo-controlled studies are needed to support these results, based on the estimated sample sizes in this study.
Collapse
Affiliation(s)
- Tomoki Takahashi
- Research and Development Department, Bean Stalk Snow Co., Ltd., Saitama 350-1165, Japan; (H.F.); (H.M.U.); (S.W.-M.); (T.N.); (T.K.)
- Correspondence: ; Tel.: +81-49-242-8138
| | - Hirofumi Fukudome
- Research and Development Department, Bean Stalk Snow Co., Ltd., Saitama 350-1165, Japan; (H.F.); (H.M.U.); (S.W.-M.); (T.N.); (T.K.)
| | - Hiroshi M. Ueno
- Research and Development Department, Bean Stalk Snow Co., Ltd., Saitama 350-1165, Japan; (H.F.); (H.M.U.); (S.W.-M.); (T.N.); (T.K.)
| | - Shiomi Watanabe-Matsuhashi
- Research and Development Department, Bean Stalk Snow Co., Ltd., Saitama 350-1165, Japan; (H.F.); (H.M.U.); (S.W.-M.); (T.N.); (T.K.)
| | - Taku Nakano
- Research and Development Department, Bean Stalk Snow Co., Ltd., Saitama 350-1165, Japan; (H.F.); (H.M.U.); (S.W.-M.); (T.N.); (T.K.)
| | - Toshiya Kobayashi
- Research and Development Department, Bean Stalk Snow Co., Ltd., Saitama 350-1165, Japan; (H.F.); (H.M.U.); (S.W.-M.); (T.N.); (T.K.)
| | - Kayoko Ishimaru
- Department of Immunology, Faculty of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan; (K.I.); (A.N.)
| | - Atsuhito Nakao
- Department of Immunology, Faculty of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan; (K.I.); (A.N.)
| |
Collapse
|
43
|
Host-microbiome intestinal interactions during early life: considerations for atopy and asthma development. Curr Opin Allergy Clin Immunol 2021; 20:138-148. [PMID: 32004178 DOI: 10.1097/aci.0000000000000629] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW The body's largest microbial community, the gut microbiome, is in contact with mucosal surfaces populated with epithelial, immune, endocrine and nerve cells, all of which sense and respond to microbial signals. These mutual interactions have led to a functional coevolution between the microbes and human physiology. Examples of coadaptation are anaerobes Bifidobacteria and Bacteroides, which have adjusted their metabolism to dietary components of human milk, and infant immune development, which has evolved to become reliant on the presence of beneficial microbes. Current research suggests that specific composition of the early-life gut microbiome aligns with the maturation of host immunity. Disruptions of natural microbial succession patterns during gut colonization are a consistent feature of immune-mediated diseases, including atopy and asthma. RECENT FINDINGS Here, we catalog recent birth cohorts documenting associations between immune dysregulation and microbial alterations, and summarize the evidence supporting the role of the gut microbiome as an etiological determinant of immune-mediated allergic diseases. SUMMARY Ecological concepts that describe microbial dynamics in the context of the host environment, and a portray of immune and neuroendocrine signaling induced by host-microbiome interactions, have become indispensable in describing the molecular role of early-life microbiome in atopy and asthma susceptibility.
Collapse
|
44
|
Yoon W, Park SH, Lee JS, Byeon JH, Kim SH, Lim J, Yoo Y. Probiotic mixture reduces gut inflammation and microbial dysbiosis in children with atopic dermatitis. Australas J Dermatol 2021; 62:e386-e392. [PMID: 34110005 DOI: 10.1111/ajd.13644] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 05/15/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Recent data suggested that dysbiosis of the gut microbiome is associated with childhood allergic diseases. Oral administration of probiotic formulations may improve the severity of atopic dermatitis (AD) by restoring imbalanced gut microbiota and reducing intestinal inflammation in children. OBJECTIVES The aim of this study was to investigate the effects of a probiotic mixture on the clinical severity of AD, gut inflammatory markers and alterations in microbiome dysbiosis in children with AD. METHODS A total of 25 subjects were enrolled in this study and administered with a mixture of probiotic strains consisting of Lactobacilli and Bifidobacteria for 4 weeks. The clinical efficacy of the probiotic mixture was assessed using SCORAD index and TEWL. Faecal calprotectin levels were measured as a marker for intestinal inflammation. The composition and diversity of the gut microbiome were analysed using 16S rRNA pyrosequencing. RESULTS The SCORAD (38.9 ± 17.2 vs 29.0 ± 15.4, P < 0.001) and TEWL (58.3 ± 12.5 vs 27.3 ± 8.7 g/m2 /h, P = 0.028) were significantly decreased after 4 weeks administration of the probiotic mixture. The faecal calprotectin level (121.5 [27.7-292.9] vs 37.0 μg/g [12.6-108.9 μg/g], P = 0.038) was significantly decreased. The α-diversity and composition of the gut microbiome were not significantly changed, but β-diversity was increased after 4 weeks. CONCLUSIONS The oral administration of the probiotic mixture was effective in reducing clinical severity and intestinal inflammation in children with AD. Gut microbial diversity was slightly increased after administration of the probiotic mixture. The results of this study suggest that a probiotic mixture can alleviate AD by decreasing inflammation and modulating the gut microbiota in children with AD.
Collapse
Affiliation(s)
- Wonsuck Yoon
- Allergy Immunology Center, Korea University, Seoul, Korea.,Environmental Health Center, Korea University Anam Hospital, Seoul, Korea
| | - Sang Hyun Park
- Department of Pediatrics, Korea University College of Medicine, Seoul, Korea
| | - Jue Seong Lee
- Department of Pediatrics, Korea University College of Medicine, Seoul, Korea
| | - Jung Hye Byeon
- Department of Pediatrics, Korea University College of Medicine, Seoul, Korea
| | - Seung Hyun Kim
- Allergy Immunology Center, Korea University, Seoul, Korea.,Environmental Health Center, Korea University Anam Hospital, Seoul, Korea
| | - Jaehoon Lim
- Allergy Immunology Center, Korea University, Seoul, Korea.,Environmental Health Center, Korea University Anam Hospital, Seoul, Korea
| | - Young Yoo
- Allergy Immunology Center, Korea University, Seoul, Korea.,Environmental Health Center, Korea University Anam Hospital, Seoul, Korea.,Department of Pediatrics, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
45
|
Szklany K, Kraneveld AD, Tiemessen MM, Garssen J, Knippels LMJ. Nutritional Interventions to Prevent the Development of Atopic Diseases: A Focus on Cow's Milk Allergy. Handb Exp Pharmacol 2021; 268:471-486. [PMID: 34085122 DOI: 10.1007/164_2021_480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
In the western world the prevalence of atopic diseases such as food allergies is increasing highly significantly. One of the earliest and most prevalent food allergies occurring in the first year of life is cow's milk allergy. No treatment is available and only avoidance of the cow's milk allergens prevents the occurrence of an allergic reaction. Since cow's milk allergic children have an increased risk of developing other allergies later in life, investigating nutritional strategies to prevent the development of cow's milk allergy by developing oral tolerance is of high interest. Nutritional components such as prebiotics, probiotics, synbiotics and long-chain polyunsaturated fatty acids possess potential to support the maturation of the immune system early in life that might prevent the development of cow's milk allergy. The available research, so far, shows promising results particularly on the development of eczema. However, the preventive effects of the nutritional interventions on the development of food allergy are inconclusive. Future research may benefit from the combination of various dietary components. To clarify the preventive effects of the nutritional components in food allergy more randomized clinical trials are needed.
Collapse
Affiliation(s)
- Kirsten Szklany
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Faculty of science, Utrecht University, Utrecht, The Netherlands
| | - Aletta D Kraneveld
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Faculty of science, Utrecht University, Utrecht, The Netherlands.,Veterinary Pharmacology and Therapeutics, Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | - Machteld M Tiemessen
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Faculty of science, Utrecht University, Utrecht, The Netherlands.,Department of Immunology, Danone Nutricia Research, Utrecht, The Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Faculty of science, Utrecht University, Utrecht, The Netherlands.,Department of Immunology, Danone Nutricia Research, Utrecht, The Netherlands
| | - Leon M J Knippels
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Faculty of science, Utrecht University, Utrecht, The Netherlands. .,Department of Immunology, Danone Nutricia Research, Utrecht, The Netherlands.
| |
Collapse
|
46
|
Montazeri-Najafabady N, Ghasemi Y, Dabbaghmanesh MH, Ashoori Y, Talezadeh P, Koohpeyma F, Abootalebi SN, Gholami A. Exploring the bone sparing effects of postbiotics in the post-menopausal rat model. BMC Complement Med Ther 2021; 21:155. [PMID: 34049521 PMCID: PMC8161980 DOI: 10.1186/s12906-021-03327-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 05/13/2021] [Indexed: 12/20/2022] Open
Abstract
Background Post-menopausal osteoporosis is a concern of health organizations, and current treatments do not seem enough. Postbiotics as bioactive compounds produced by probiotics may be an attractive alternative for bone health. In this study, we prepared, formulated, and compared the effects of cell lysate and supernatant of five native probiotic strains (Lactobacillus acidophilus, Lactobacillus reuteri, Lactobacillus casei, Bifidobacterium longum, and Bacillus coagulans) in ovariectomized (OVX) rats. Methods The probiotic strains were isolated, and their cell-free supernatants and biomasses as postbiotics were extracted and formulated using standard microbial processes. The Sprague-Dawley rats were fed by 1 × 109 CFU/ml/day postbiotic preparations for 4 weeks immediately after ovariectomy. Dual-energy X-ray absorptiometry (DEXA) scans were accomplished to evaluate femur, spine, and tibia BMD. The serum biochemical markers [calcium, phosphorus, and alkaline phosphatase] were assessed. Results Postbiotics could considerably improve the global and femur area in OVX rats. In the case of global bone mineral density (BMD), Lactobacillus casei lysate and supernatant, Bacillus coagulans lysate and supernatant, lysate of Bifidobacterium longum and Lactobacillus acidophilus, and Lactobacillus reuteri supernatant significantly increased BMD. We found Bacillus coagulans supernatant meaningfully enriched tibia BMD. Conclusion Postbiotic could ameliorate bone loss resulting from estrogen deficiency. Also, the effects of postbiotics on different bone sites are strain-dependent. More clinical studies need to explore the optimal administrative dose and duration of the specific postbiotics in protecting bone loss.
Collapse
Affiliation(s)
- Nima Montazeri-Najafabady
- Endocrinology and Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Biotechnology Research Center, Shiraz University of Medical Sciences, P.O. Box: 71348-14336, Shiraz, Iran
| | - Younes Ghasemi
- Pharmaceutical Science Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Yousef Ashoori
- Pharmaceutical Science Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Pedram Talezadeh
- Endocrinology and Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farhad Koohpeyma
- Endocrinology and Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyedeh Narjes Abootalebi
- Division of Intensive Care Unit, Department of Pediatrics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Gholami
- Biotechnology Research Center, Shiraz University of Medical Sciences, P.O. Box: 71348-14336, Shiraz, Iran.
| |
Collapse
|
47
|
Sun S, Chang G, Zhang L. The prevention effect of probiotics against eczema in children: an update systematic review and meta-analysis. J DERMATOL TREAT 2021; 33:1844-1854. [PMID: 34006167 DOI: 10.1080/09546634.2021.1925077] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Accumulated evidences support the fetus's intestinal flora unbalance is associated with the development of allergic diseases. Probiotic supplements in pregnancy and childhood might prevent atopic diseases. The aim of this systematic review and meta-analysis was to evaluate the effect of probiotic supplementation during pregnancy and early infancy in preventing eczema, atopic eczema, and other allergic diseases. We also explored whether different probiotic strains or intervention objects affected the antiallergic effect of probiotics and the prevention atopy effect of the long-term period. Fixed-effect models were used, and random-effects models where significant heterogeneity was present. Results were expressed as odds ratios (ORs) with a 95% confidence interval (CI). Twenty-one studies were included in the meta-analysis. The probiotics group had a significantly lower risk of eczema and atopic eczema compared to controls, especially those treated with probiotic combinations. Mothers' probiotics intake significantly contributed to reducing the risk of eczema as well as atopic eczema. What's more, probiotics seemed effective on eczema prevention ≤2 years of age, but against atopic eczema after 1 of age year. No significant difference in terms of prevention of asthma, rhinitis, wheeze, allergic diseases and sensation. In brief, a probiotic supplement is expected to become a novel potential strategy for infant eczema and atopic eczema.
Collapse
Affiliation(s)
- Shuya Sun
- Graduate school, Tianjin Medical University, Tianjin, China
| | - Guizhen Chang
- Department of Dermatology, Tianjin TEDA Hospital, Tianjin, China
| | - Litao Zhang
- Department of Dermatology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
| |
Collapse
|
48
|
Chiu CJ, Huang MT. Asthma in the Precision Medicine Era: Biologics and Probiotics. Int J Mol Sci 2021; 22:4528. [PMID: 33926084 PMCID: PMC8123613 DOI: 10.3390/ijms22094528] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/23/2021] [Accepted: 04/24/2021] [Indexed: 02/07/2023] Open
Abstract
Asthma is a major global health issue. Over 300 million people worldwide suffer from this chronic inflammatory airway disease. Typical clinical symptoms of asthma are characterized by a recurrent wheezy cough, chest tightness, and shortness of breath. The main goals of asthma management are to alleviate asthma symptoms, reduce the risk of asthma exacerbations, and minimize long-term medicinal adverse effects. However, currently available type 2 T helper cells (Th2)-directed treatments are often ineffective due to the heterogeneity of the asthma subgroups, which manifests clinically with variable and poor treatment responses. Personalized precision therapy of asthma according to individualized clinical characteristics (phenotype) and laboratory biomarkers (endotype) is the future prospect. This mini review discusses the molecular mechanisms underlying asthma pathogenesis, including the hot sought-after topic of microbiota, add-on therapies and the potential application of probiotics in the management of asthma.
Collapse
Affiliation(s)
- Chiao-Juno Chiu
- Graduate Institute of Clinical Medicine, School of Medicine, National Taiwan University, Taipei 100, Taiwan;
| | - Miao-Tzu Huang
- Graduate Institute of Clinical Medicine, School of Medicine, National Taiwan University, Taipei 100, Taiwan;
- Department of Medical Research, National Taiwan University Hospital, No. 7 Chung-Shan South Road, Taipei 100, Taiwan
- Department of Pediatrics, National Taiwan University Hospital, No. 7 Chung-Shan South Road, Taipei 100, Taiwan
| |
Collapse
|
49
|
Sun M, Luo J, Liu H, Xi Y, Lin Q. Can Mixed Strains of Lactobacillus and Bifidobacterium Reduce Eczema in Infants under Three Years of Age? A Meta-Analysis. Nutrients 2021; 13:nu13051461. [PMID: 33923096 PMCID: PMC8145948 DOI: 10.3390/nu13051461] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/19/2021] [Accepted: 04/22/2021] [Indexed: 11/16/2022] Open
Abstract
(1) Background: Whether early supplementation of probiotics to improve intestinal flora can effectively prevent eczema remains a controversial issue. We aimed to investigate the effect of a mixed strain of Lactobacillus and Bifidobacterium on eczema in infants under three years old at present; (2) Methods: We searched the databases of PubMed, Web of Science, and Cochrane Library, as well as National Knowledge Infrastructure (CNKI), WeiPu (VIP), and WanFang Data (WanFang) for randomized controlled trials (RCTs) of probiotics in the prevention of eczema in infants without language restriction. The main outcome was eczema incidence, while adverse events during the intervention constituted the secondary outcome. The random-/fixed-effects model was utilized to calculate the combined relative risk (RR) and 95% confidence interval (CI). The methodological quality of the study was evaluated using the Cochrane "bias risk" tool. According to the initial intervention time, subgroup analysis was carried out, follow-up time, family history, etc.; (3) Results: Nine articles were selected (2093 infants). The Lactobacillus and Bifidobacterium mixed strain could prevent eczema in infants under three years of age compared to the placebo (RR = 0.60; I2 = 67%; p < 0.001). Subgroup analysis revealed that the mixture of two probiotic strains had preventive effects on both infants with positive (RR = 0.53; I2 = 52%; p < 0.001) and negative (RR = 0.69; I2 = 62%; p = 0.02) family history; The follow-up time for ≤12 months (RR = 0.65; I2 = 12%; p = 0.01) and 12-24 months (RR = 0.60; I2 = 79%; p = 0.003), daily dose of probiotics ≤ 1 × 109 and > 1 × 109 colony forming units all can be effective (p < 0.01); Compared with the intervention of infants alone (RR = 0.63; I2 = 63%; p = 0.29), the effect of probiotics mixture at the beginning of pregnancy was more significant (RR = 0.59; I2 = 71%; p < 0.001); Except for the mixture of Lactobacillus rhamnosusGG (LGG) and Bifidobacterium longum (B. longum) (p = 0.18), other subgroups of intervention group can play a preventive effect (p < 0.05); (4) Conclusions: The mixed strain of Lactobacillus and Bifidobacterium can effectively reduce the incidence of eczema in infants under three years old. However, further research is needed to fully understand the exact mechanism of their effect on infant eczema.
Collapse
Affiliation(s)
| | | | | | | | - Qian Lin
- Correspondence: ; Tel.: +86-0731-82650291
| |
Collapse
|
50
|
Carucci L, Coppola S, Luzzetti A, Giglio V, Vanderhoof J, Berni Canani R. The role of probiotics and postbiotics in modulating the gut microbiome-immune system axis in the pediatric age. Minerva Pediatr (Torino) 2021; 73:115-127. [PMID: 33880903 DOI: 10.23736/s2724-5276.21.06188-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The complex microbial community of the gut microbiome plays a fundamental role in driving development and function of the human immune system. This phenomenon is named the gut microbiome-immune system axis. When operating optimally, this axis influences both innate and adaptive immunity, which orchestrates the maintenance of crucial elements of host-microorganisms symbiosis, in a dialogue that modulates responses in the most beneficial way. Growing evidence reveals some environmental factors which can positively and negatively modulate the gut microbiome-immune system axis with consequences on the body health status. Several conditions which increasingly affect the pediatric age, such as allergies, autoimmune and inflammatory disorders, arise from a failure of the gut microbiome-immune system axis. Prenatal or postnatal modulation of this axis through some interventional strategies (including diet, probiotics and postbiotics), may lead to a positive gene-environment interaction with improvement of immune-modulatory effects and final positive effect on human health. In particular probiotics and postbiotics exerting pleiotropic regulatory actions on the gut-microbiome-immune system axis provide an innovative preventive and therapeutic strategy for many pediatric conditions.
Collapse
Affiliation(s)
- Laura Carucci
- Department of Translational Medical Science, Federico II University, Naples, Italy.,ImmunoNutritionLab at CEINGE Advanced Biotechnologies, Federico II University, Naples, Italy
| | - Serena Coppola
- Department of Translational Medical Science, Federico II University, Naples, Italy.,ImmunoNutritionLab at CEINGE Advanced Biotechnologies, Federico II University, Naples, Italy
| | - Anna Luzzetti
- Department of Translational Medical Science, Federico II University, Naples, Italy.,ImmunoNutritionLab at CEINGE Advanced Biotechnologies, Federico II University, Naples, Italy
| | - Veronica Giglio
- Department of Translational Medical Science, Federico II University, Naples, Italy.,ImmunoNutritionLab at CEINGE Advanced Biotechnologies, Federico II University, Naples, Italy
| | - Jon Vanderhoof
- Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Roberto Berni Canani
- Department of Translational Medical Science, Federico II University, Naples, Italy - .,ImmunoNutritionLab at CEINGE Advanced Biotechnologies, Federico II University, Naples, Italy.,European Laboratory for the Investigation of Food-Induced Diseases, Federico II University, Naples, Italy.,Task Force for Microbiome Studies, Federico II University, Naples, Italy
| |
Collapse
|