1
|
Wang HZ, Hayles EH, Fiander M, Sinn JK, Osborn DA. Probiotics in infants for prevention of allergic disease. Cochrane Database Syst Rev 2025; 6:CD006475. [PMID: 40511642 PMCID: PMC12163975 DOI: 10.1002/14651858.cd006475.pub3] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/16/2025]
Abstract
RATIONALE This is an update of a Cochrane review first published in 2007. Allergic disease and food allergy are prevalent, and contribute to a significant burden of disease on the individual, their family and the healthcare system. Probiotics are live bacteria that colonise the gastrointestinal tract, and have been studied in many clinical trials for preventing allergic conditions. OBJECTIVES To evaluate the benefits and harms of a probiotic, or a probiotic with added prebiotic ('synbiotic'), compared with control (placebo or no treatment) for preventing allergic diseases (asthma, eczema, allergic rhinitis) and dietary allergies in infants by two years of age. SEARCH METHODS We searched CENTRAL, MEDLINE, Embase and trial registries in December 2023. We reviewed the reference lists of studies selected for inclusion in this review, and systematic reviews on similar topics. We manually searched conference abstracts. ELIGIBILITY CRITERIA We included randomised controlled trials that compared a probiotic to a control, or a probiotic added to a prebiotic ('synbiotic'). We included enterally fed infants in the first six months of life without clinical evidence of allergic disease. We included probiotics added to human milk or infant formula, added in the manufacturing process or given separately. OUTCOMES Infant incidence by two years of age and childhood incidence (up to 10 years of age or up to the age of latest report between 2 and 10 years) of specific allergic diseases, including: asthma, eczema, allergic rhinitis, immunoglobulin E (IgE)-mediated food allergy, IgE-mediated cow's milk protein allergy. Events of anaphylaxis and potential harms including adverse effects, harms or infection with probiotic bacteria. RISK OF BIAS We used the Cochrane RoB 2 tool to assess bias in the studies. SYNTHESIS METHODS We used the random-effects (Mantel-Haenszel) model for meta-analysis where possible. Where this was not possible due to the nature of the data, we synthesised and interpreted individual studies separately. We used GRADE to assess the certainty of evidence for each outcome. INCLUDED STUDIES We included 24 studies (7077 mother-infant pairs). The studies were conducted in many parts of the world, including the USA, Europe, South Korea, Japan, Singapore and Australia, with most being conducted in Europe. Studies were published between 2001 and 2020. As some studies measured outcomes such as eczema using different criteria, we made assumptions to allow us to combine data. SYNTHESIS OF RESULTS Probiotics may result in little to no difference in asthma (risk ratio (RR) 0.96, 95% confidence interval (CI) 0.65 to 1.44; 4 studies, 954 participants; low-certainty evidence), allergic rhinitis (RR 0.89, 95% CI 0.45 to 1.77; 5 studies, 1045 participants; low-certainty evidence) and IgE-mediated cow's milk protein allergy (RR 0.99, 95% CI 0.82 to 1.20; 4 studies, 259 participants; low-certainty evidence) by two years of age. Probiotics may result in a slight reduction in eczema by two years of age (RR 0.87, 95% CI 0.78 to 0.97; 18 studies, 3494 participants; low-certainty evidence); however, sensitivity analysis of the studies at low risk of bias showed little or no difference in eczema by two years of age (RR 0.86, 95% CI 0.69 to 1.07; 4 studies, 892 participants). Probiotic supplementation may have little to no effect on the incidence of food allergy by two years, but the evidence is very uncertain (RR 1.12, 95% CI 0.57 to 2.20; 3 studies, 857 participants; very low-certainty evidence). The evidence is very uncertain about the effect of synbiotics on eczema by two years of age (RR 0.88, 95% CI 0.52 to 1.47; 3 studies, 1235 participants; very low-certainty evidence). Synbiotics may result in little to no difference in food allergy by two years of age (RR 1.06, 95% CI 0.55 to 2.07; 1 study, 223 participants; low-certainty evidence). There were no data for the effect of synbiotics on asthma, allergic rhinitis and IgE-mediated cow's milk protein allergy by two years of age. Probiotic or synbiotic supplementation may result in little to no difference in potential harms including adverse effects, harms or infection with probiotic bacteria at any point during the study intervention by two years of age. There were no serious adverse events related to probiotics or synbiotics reported. We had some concerns about risk of bias for most studies, with only a few judged at low risk of bias. Some studies had a high risk of bias due to unclear randomisation, missing data and lack of prespecified intentions. Estimates were often imprecise, with wide CIs due to limited events. The limited data prevented subgroup analyses on infant risk factors and feeding methods for outcomes other than the effect of probiotics on eczema. Only three studies assessed synbiotic supplementation, leaving their role in allergic disease prevention uncertain. The included studies were mainly in high-income countries in many different areas of the world, but may have limited applicability to other regions. AUTHORS' CONCLUSIONS There is insufficient evidence to make conclusions about the effect of probiotics and synbiotics on preventing the development of allergic diseases by two years of age and during childhood up to 10 years of age. Although there were no serious adverse events reported for the use of probiotics in infants, incorporating probiotics and synbiotics into routine practice requires further information to support their use. FUNDING This Cochrane review had no dedicated funding. REGISTRATION Protocol (2007) available via https://doi.org/10.1002/14651858.CD006475. Original review (2007) available via https://doi.org/10.1002/14651858.CD006475.pub2.
Collapse
Affiliation(s)
- Hang Zhen Wang
- Macquarie Medical School, Macquarie University, Sydney, Australia
| | - Elizabeth H Hayles
- Department of Neonatology, Royal North Shore Hospital, The University of Sydney, St Leonards, Australia
| | | | - John Kh Sinn
- Department of Neonatology, Royal North Shore Hospital, Sydney, Australia
- Faculty of Medicine and Health Sciences, Macquarie University, Macquarie University, Sydney, Australia
| | - David A Osborn
- Central Clinical School, School of Medicine, The University of Sydney, Sydney, Australia
| |
Collapse
|
2
|
Finnegan YE, Neill HR, Prpa EJ, Pot B. "Gut" to grips with the science of the microbiome - a symposium report. GUT MICROBIOME (CAMBRIDGE, ENGLAND) 2024; 5:e11. [PMID: 39703540 PMCID: PMC11658944 DOI: 10.1017/gmb.2024.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 06/18/2024] [Indexed: 12/21/2024]
Abstract
The latest Yakult Science Study Day was held virtually on 2 November 2023. Aimed at healthcare professionals, researchers, and students, a variety of experts explored the latest gut microbiome research and what it means in practice. The morning sessions discussed the role of the microbiome in health and disease, the rapid advancements in DNA sequencing and implications for personalised nutrition, the current state of evidence on health benefits associated with fermented foods, prebiotics and probiotics and the challenges involved in interpreting research in this area. The afternoon session considered the emerging research on the microbiota-gut-brain axis in mediating effects of food on mood, the bidirectional impact of menopause on the gut microbiota, and the interplay between the gut and skin with implications for the treatment of rare and common skin disorders. The session ended with an update on the use of faecal microbiota transplant in both research and clinical practice. Undoubtedly, the gut microbiome is emerging as a key conductor of human health, both in relation to gastrointestinal and non-gastrointestinal outcomes. As research continues to elucidate mechanisms of action and confirm their effects in human trials, the gut microbiome should be a key consideration within a holistic approach to health moving forward.
Collapse
Affiliation(s)
- Yvonne E. Finnegan
- Yvonne Finnegan FINNE Nutrition & Regulatory Consultancy, Kilkenny, Ireland
| | | | | | - Bruno Pot
- Yakult Europe BV, Science Department, Almere, The Netherlands
| |
Collapse
|
3
|
Wang S, Yin P, Yu L, Tian F, Chen W, Zhai Q. Effects of Early Diet on the Prevalence of Allergic Disease in Children: A Systematic Review and Meta-Analysis. Adv Nutr 2024; 15:100128. [PMID: 37827490 PMCID: PMC10831899 DOI: 10.1016/j.advnut.2023.10.001] [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: 02/23/2023] [Revised: 08/08/2023] [Accepted: 10/06/2023] [Indexed: 10/14/2023] Open
Abstract
Recent evidence suggests that the timing of introduction, types, and amounts of complementary foods/allergenic foods may influence the risk of allergic disease. However, the evidence has not been updated and comprehensively synthesized. The Cochrane Library, EMBASE, Web of Science, and PubMed databases were searched from the inception of each database up to 31 May 2023 (articles prior to 2000 were excluded manually). Statistical analyses were performed using RevMan 5. The GRADE approach was followed to rate the certainty of evidence. Compared with >6 mo, early introduction of eggs (≤6 mo of age) might reduce the risk of food allergies in preschoolers aged <6 y (odds ratio [OR], 0.65; 95% confidence interval [CI], 0.53, 0.81), but had no effect on asthma or atopic dermatitis (AD). Consumption of fish at 6-12 mo might reduce the risk of asthma in children (aged 5-17 y) compared with late introduction after 12 mo (OR, 0.61; 95% CI: 0.52, 0.72). Introduction of allergenic foods for ≤6 mo of age, compared with >6 mos, was a protective factor for the future risk (children aged ≤10 y) of AD (OR, 0.93; 95% CI: 0.89, 0.97). Probiotic intervention for infants at high risk of allergic disease significantly reduced the risk of food allergy at ages 0-3 y (OR, 0.72; 95% CI: 0.56, 0.94), asthma at 6-12 y (OR, 0.61; 95% CI: 0.41, 0.90), and AD at aged <6 y (3-6 y: OR, 0.70; 95% CI: 0.52, 0.94; 0-3 y: OR, 0.73; 95% CI: 0.59, 0.91). Early introduction of complementary foods or the high-dose vitamin D supplementation in infancy was not associated with the risk of developing food allergies, asthma, or AD during childhood. Early introduction to potential allergen foods for normal infants or probiotics for infants at high risk of allergies may protect against development of allergic disease. This study was registered at PROSPERO as CRD42022379264.
Collapse
Affiliation(s)
- Shumin Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Pingping Yin
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Leilei Yu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China.
| |
Collapse
|
4
|
Radford-Smith DE, Anthony DC. Prebiotic and Probiotic Modulation of the Microbiota-Gut-Brain Axis in Depression. Nutrients 2023; 15:nu15081880. [PMID: 37111100 PMCID: PMC10146605 DOI: 10.3390/nu15081880] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 04/07/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Emerging evidence demonstrates that alterations to the gut microbiota can affect mood, suggesting that the microbiota-gut-brain (MGB) axis contributes to the pathogenesis of depression. Many of these pathways overlap with the way in which the gut microbiota are thought to contribute to metabolic disease progression and obesity. In rodents, prebiotics and probiotics have been shown to modulate the composition and function of the gut microbiota. Together with germ-free rodent models, probiotics have provided compelling evidence for a causal relationship between microbes, microbial metabolites, and altered neurochemical signalling and inflammatory pathways in the brain. In humans, probiotic supplementation has demonstrated modest antidepressant effects in individuals with depressive symptoms, though more studies in clinically relevant populations are needed. This review critically discusses the role of the MGB axis in depression pathophysiology, integrating preclinical and clinical evidence, as well as the putative routes of communication between the microbiota-gut interface and the brain. A critical overview of the current approaches to investigating microbiome changes in depression is provided. To effectively translate preclinical breakthroughs in MGB axis research into novel therapies, rigorous placebo-controlled trials alongside a mechanistic and biochemical understanding of prebiotic and probiotic action are required from future research.
Collapse
Affiliation(s)
- Daniel E Radford-Smith
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
- Department of Chemistry, University of Oxford, Mansfield Road, Oxford OX1 3TA, UK
- Department of Psychiatry, University of Oxford, Warneford Hospital, Warneford Lane, Oxford OX3 7JX, UK
| | - Daniel C Anthony
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| |
Collapse
|
5
|
Rahman T, Sarwar PF, Potter C, Comstock SS, Klepac-Ceraj V. Role of human milk oligosaccharide metabolizing bacteria in the development of atopic dermatitis/eczema. Front Pediatr 2023; 11:1090048. [PMID: 37020647 PMCID: PMC10069630 DOI: 10.3389/fped.2023.1090048] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 02/23/2023] [Indexed: 04/07/2023] Open
Abstract
Despite affecting up to 20% of infants in the United States, there is no cure for atopic dermatitis (AD), also known as eczema. Atopy usually manifests during the first six months of an infant's life and is one predictor of later allergic health problems. A diet of human milk may offer protection against developing atopic dermatitis. One milk component, human milk oligosaccharides (HMOs), plays an important role as a prebiotic in establishing the infant gut microbiome and has immunomodulatory effects on the infant immune system. The purpose of this review is to summarize the available information about bacterial members of the intestinal microbiota capable of metabolizing HMOs, the bacterial genes or metabolic products present in the intestinal tract during early life, and the relationship of these genes and metabolic products to the development of AD/eczema in infants. We find that specific HMO metabolism gene sets and the metabolites produced by HMO metabolizing bacteria may enable the protective role of human milk against the development of atopy because of interactions with the immune system. We also identify areas for additional research to further elucidate the relationship between the human milk metabolizing bacteria and atopy. Detailed metagenomic studies of the infant gut microbiota and its associated metabolomes are essential for characterizing the potential impact of human milk-feeding on the development of atopic dermatitis.
Collapse
Affiliation(s)
- Trisha Rahman
- Department of Biological Sciences, Wellesley College, Wellesley, MA, United States
| | - Prioty F. Sarwar
- Department of Biological Sciences, Wellesley College, Wellesley, MA, United States
| | - Cassie Potter
- Department of Biological Sciences, Wellesley College, Wellesley, MA, United States
| | - Sarah S. Comstock
- Department of Food Science & Human Nutrition, Michigan State University, East Lansing, MI, United States
| | - Vanja Klepac-Ceraj
- Department of Biological Sciences, Wellesley College, Wellesley, MA, United States
| |
Collapse
|
6
|
Xie A, Chen A, Chen Y, Luo Z, Jiang S, Chen D, Yu R. Lactobacillus for the treatment and prevention of atopic dermatitis: Clinical and experimental evidence. Front Cell Infect Microbiol 2023; 13:1137275. [PMID: 36875529 PMCID: PMC9978199 DOI: 10.3389/fcimb.2023.1137275] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease, accompanied by itching and swelling. The main pathological mechanism of AD is related to the imbalance between Type 2 helper cells (Th2 cells) and Type 1 helper cells (Th1 cells). Currently, no safe and effective means to treat and prevent AD are available; moreover, some treatments have side effects. Probiotics, such as some strains of Lactobacillus, can address these concerns via various pathways: i) facilitating high patient compliance; ii) regulating Th1/Th2 balance, increasing IL-10 secretion, and reducing inflammatory cytokines; iii) accelerating the maturation of the immune system, maintaining intestinal homeostasis, and improving gut microbiota; and iv) improving the symptoms of AD. This review describes the treatment and prevention of AD using 13 species of Lactobacillus. AD is commonly observed in children. Therefore, the review includes a higher proportion of studies on AD in children and fewer in adolescents and adults. However, there are also some strains that do not improve the symptoms of AD and even worsen allergies in children. In addition, a subset of the genus Lactobacillus that can prevent and relieve AD has been identified in vitro. Therefore, future studies should include more in vivo studies and randomized controlled clinical trials. Given the advantages and disadvantages mentioned above, further research in this area is urgently required.
Collapse
Affiliation(s)
- Anni Xie
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Ailing Chen
- Research Institute for Reproductive Health and Genetic Diseases, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yuqing Chen
- Department of Child Health Care, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Zichen Luo
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Shanyu Jiang
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Daozhen Chen
- Research Institute for Reproductive Health and Genetic Diseases, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
- *Correspondence: Daozhen Chen, ; Renqiang Yu,
| | - Renqiang Yu
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Research Institute for Reproductive Health and Genetic Diseases, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
- *Correspondence: Daozhen Chen, ; Renqiang Yu,
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
Dekker J, Quilter M, Qian H. Comparison of two probiotics in follow-on formula: Bifidobacterium animalis subsp. lactis HN019 reduced upper respiratory tract infections in Chinese infants. Benef Microbes 2022; 13:341-354. [PMID: 36004715 DOI: 10.3920/bm2022.0041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A randomised, double-blind, placebo-controlled trial was performed to investigate the health benefits of probiotic bacteria in infants when delivered in a follow-on infant formula. The study was conducted in Fuyang (Anhui Province, China) during winter and enrolled 192 healthy infants aged six to 12 months. Infants received one of three follow-on formulae daily for 12 weeks: supplemented with 106 cfu/g Bifidobacterium animalis subsp. lactis HN019 (n=64); 106 cfu/g Lacticaseibacillus rhamnosus HN001 (n=64); or without added probiotics (n=64). The primary endpoint was physician-confirmed bacterial or viral infections during the treatment period. Secondary endpoints included parentally reported (confirmed and unconfirmed) infections; antiviral or antibiotic treatments, and hospitalisation; stool frequency and consistency; infant growth; infant temperament; and adverse events. There were 8 cases of confirmed infection, all upper respiratory tract infections (URTIs). Confirmed URTIs were observed in 9.4% of the control group, compared to 3.1% in the HN001 group (P=0.273), and 0.0% in the HN019 group (P=0.028). A similar trend was observed for parentally reported URTIs, with 25.0% in the control group, compared with 14.1% in the HN001 group (P=0.119) and 9.4% in the HN019 group (P=0.019). No infants in the HN019 group were prescribed antibiotics or antivirals, compared with 3 (4.7%) in the HN001 group and 7 (10.9%) in the control group. No infants required hospitalisation. The probiotic-containing formulae were well-tolerated: there were no cases of diarrhoea or differences in stool frequency or characteristics, no differences in infant growth or temperament, and no treatment-related adverse events. This study directly compared the benefits of two different probiotics when added to follow-on infant formula at 106 cfu/g and consumed over a 12-week period. While HN001 showed trends toward reduced infections, HN019 showed better performance in terms of significantly reduced incidence of both physician-confirmed and parentally reported URTIs, and antibiotic/antiviral use compared to a control in Chinese infants. The trial is registered at ClinicalTrials.gov (NCT01724203).
Collapse
Affiliation(s)
- J Dekker
- Fonterra Research and Development Centre, Palmerston North, Private Bag 11029, 4442 Palmerston North, New Zealand
| | - M Quilter
- Fonterra Research and Development Centre, Palmerston North, Private Bag 11029, 4442 Palmerston North, New Zealand
| | - H Qian
- School of Public Health, Anhui Medical University, Hefei, China P.R
| |
Collapse
|
10
|
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: 10] [Impact Index Per Article: 3.3] [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
|
11
|
Ciprandi G, Tosca MA. Probiotics in Children with Asthma. CHILDREN (BASEL, SWITZERLAND) 2022; 9:978. [PMID: 35883962 PMCID: PMC9316460 DOI: 10.3390/children9070978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 06/23/2022] [Accepted: 06/27/2022] [Indexed: 12/11/2022]
Abstract
A type-2 immune response usually sustains wheezing and asthma in children. In addition, dysbiosis of digestive and respiratory tracts is detectable in patients with wheezing and asthma. Probiotics may rebalance immune response, repair dysbiosis, and mitigate airway inflammation. As a result, probiotics may prevent asthma and wheezing relapse. There is evidence that some probiotic strains may improve asthma outcomes in children. In this context, the PROPAM study provided evidence that two specific strains significantly prevented asthma exacerbations and wheezing episodes. Therefore, oral probiotics could be used as add-on asthma therapy in managing children with asthma, but the choice should be based on documented evidence.
Collapse
Affiliation(s)
- Giorgio Ciprandi
- Allergy Clinic, Casa di Cura Villa Montallegro, 16146 Genoa, Italy
| | | |
Collapse
|
12
|
Manipulating Microbiota to Treat Atopic Dermatitis: Functions and Therapies. Pathogens 2022; 11:pathogens11060642. [PMID: 35745496 PMCID: PMC9228373 DOI: 10.3390/pathogens11060642] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 05/27/2022] [Accepted: 05/29/2022] [Indexed: 12/13/2022] Open
Abstract
Atopic dermatitis (AD) is a globally prevalent skin inflammation with a particular impact on children. Current therapies for AD are challenged by the limited armamentarium and the high heterogeneity of the disease. A novel promising therapeutic target for AD is the microbiota. Numerous studies have highlighted the involvement of the skin and gut microbiota in the pathogenesis of AD. The resident microbiota at these two epithelial tissues can modulate skin barrier functions and host immune responses, thus regulating AD progression. For example, the pathogenic roles of Staphylococcus aureus in the skin are well-established, making this bacterium an attractive target for AD treatment. Targeting the gut microbiota is another therapeutic strategy for AD. Multiple oral supplements with prebiotics, probiotics, postbiotics, and synbiotics have demonstrated promising efficacy in both AD prevention and treatment. In this review, we summarize the association of microbiota dysbiosis in both the skin and gut with AD, and the current knowledge of the functions of commensal microbiota in AD pathogenesis. Furthermore, we discuss the existing therapies in manipulating both the skin and gut commensal microbiota to prevent or treat AD. We also propose potential novel therapies based on the cutting-edge progress in this area.
Collapse
|
13
|
Association of Probiotics with Atopic Dermatitis among Infant: A Meta-analysis of Randomized Controlled Trials. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5080190. [PMID: 35651728 PMCID: PMC9150986 DOI: 10.1155/2022/5080190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/12/2022] [Accepted: 03/23/2022] [Indexed: 11/17/2022]
Abstract
Background. Previous studies have explored the relationship between probiotics and risk of atopic dermatitis among infant; however, the results are still inconclusive. We aimed to assess the abovementioned association. Methods. PubMed, Web of Science, Embase, and China National Knowledge Infrastructure were retrieved for association between probiotics and atopic dermatitis with randomized controlled trials (RCTs) until Nov 20, 2021. The effect size was pooled by using random or fixed effect models according to the heterogeneity. Stata 12.0 was used for meta-analysis, sensitivity analysis, and bias analysis. Results. At the end of the screening article, 2575 infants were extracted from 8 trials and finally met the qualification criteria. In comparison to placebo, probiotics dramatically reduced incidence of childhood atopic dermatitis (
, 95%
-0.95). However, probiotics did not exhibit benefit over placebo in preventing the development of either IgE-associated infant AD (
, 95%
-1.22) or sensitive constitution (
, 95%
-1.08). From the results of sensitivity and publication bias, we found that these results were robust with little publication bias. Conclusion. During the late stages of pregnancy, women taking probiotics could lower the risk of infantile atopic dermatitis, but not for IgE-associated infant AD or sensitive constitution. The results could provide evidence for the fibrosis. Future studies are needed to confirm the results.
Collapse
|
14
|
Skin Microbiota in Atopic Dermatitis. Int J Mol Sci 2022; 23:ijms23073503. [PMID: 35408862 PMCID: PMC8998607 DOI: 10.3390/ijms23073503] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/18/2022] [Accepted: 03/22/2022] [Indexed: 12/26/2022] Open
Abstract
The skin microbiota represents an ecosystem composed of numerous microbial species interacting with each other, as well as with host epithelial and immune cells. The microbiota provides health benefits to the host by supporting essential functions of the skin and inhibiting colonization with pathogens. However, the disturbance of the microbial balance can result in dysbiosis and promote skin diseases, such as atopic dermatitis (AD). This review provides a current overview of the skin microbiota involvement in AD and its complex interplay with host immune response mechanisms, as well as novel therapeutic strategies for treating AD focused on restoring skin microbial homeostasis.
Collapse
|
15
|
Miko E, Csaszar A, Bodis J, Kovacs K. The Maternal-Fetal Gut Microbiota Axis: Physiological Changes, Dietary Influence, and Modulation Possibilities. Life (Basel) 2022; 12:424. [PMID: 35330175 PMCID: PMC8955030 DOI: 10.3390/life12030424] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 02/07/2023] Open
Abstract
The prenatal period and the first years of life have a significant impact on the health issues and life quality of an individual. The appropriate development of the immune system and the central nervous system are thought to be major critical determining events. In parallel to these, establishing an early intestinal microbiota community is another important factor for future well-being interfering with prenatal and postnatal developmental processes. This review aims at summarizing the main characteristics of maternal gut microbiota and its possible transmission to the offspring, thereby affecting fetal and/or neonatal development and health. Since maternal dietary factors are potential modulators of the maternal-fetal microbiota axis, we will outline current knowledge on the impact of certain diets, nutritional factors, and nutritional modulators during pregnancy on offspring's microbiota and health.
Collapse
Affiliation(s)
- Eva Miko
- Department of Medical Microbiology and Immunology, Medical School, University of Pécs, 12 Szigeti Street, 7624 Pécs, Hungary
- National Laboratory for Human Reproduction, University of Pécs, 7624 Pécs, Hungary; (A.C.); (J.B.); (K.K.)
- Janos Szentagothai Research Centre, 20 Ifjusag Street, 7624 Pécs, Hungary
| | - Andras Csaszar
- National Laboratory for Human Reproduction, University of Pécs, 7624 Pécs, Hungary; (A.C.); (J.B.); (K.K.)
- Department of Obstetrics and Gynaecology, Medical School, University of Pécs, 17 Edesanyak Street, 7624 Pécs, Hungary
| | - Jozsef Bodis
- National Laboratory for Human Reproduction, University of Pécs, 7624 Pécs, Hungary; (A.C.); (J.B.); (K.K.)
- Department of Obstetrics and Gynaecology, Medical School, University of Pécs, 17 Edesanyak Street, 7624 Pécs, Hungary
| | - Kalman Kovacs
- National Laboratory for Human Reproduction, University of Pécs, 7624 Pécs, Hungary; (A.C.); (J.B.); (K.K.)
- Department of Obstetrics and Gynaecology, Medical School, University of Pécs, 17 Edesanyak Street, 7624 Pécs, Hungary
| |
Collapse
|
16
|
Cheng J, Laitila A, Ouwehand AC. Bifidobacterium animalis subsp. lactis HN019 Effects on Gut Health: A Review. Front Nutr 2022; 8:790561. [PMID: 34970580 PMCID: PMC8712437 DOI: 10.3389/fnut.2021.790561] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/22/2021] [Indexed: 12/19/2022] Open
Abstract
Optimal gut motility is central to bowel function and gut health. The link between the gut dysmotility related disorders and dysfunctional-intestinal barriers has led to a hypothesis that certain probiotics could help in normalizing gut motility and maintain gut health. This review investigates the roles of Bifidobacterium animalis subsp. lactis HN019 (B. lactis HN019™) on gut health, and its mechanisms of action in various pre-clinical and clinical studies. Research supports the hypothesis that B. lactis HN019™ has a beneficial role in maintaining intestinal barrier function during gastrointestinal infections by competing and excluding potential pathogens via different mechanisms; maintaining normal tight junction function in vitro; and regulating host immune defense toward pathogens in both in vitro and human studies. This has been observed to lead to reduced incidence of diarrhea. Interestingly, B. lactis HN019™ also supports normal physiological function in immunosenescent elderly and competes and excludes potential pathogens. Furthermore, B. lactis HN019™ reduced intestinal transit time and increased bowel movement frequency in functional constipation, potentially by modulating gut–brain–microbiota axis, mainly via serotonin signaling pathway, through short chain fatty acids derived from microbial fermentation. B. lactis HN019™ is thus a probiotic that can contribute to relieving gut dysmotility related disorders.
Collapse
Affiliation(s)
- Jing Cheng
- International Flavors & Fragrances Inc., Global Health and Nutrition Science, Danisco Sweeteners Oy, Kantvik, Finland
| | - Arja Laitila
- International Flavors & Fragrances Inc., Global Health and Nutrition Science, Danisco Sweeteners Oy, Kantvik, Finland
| | - Arthur C Ouwehand
- International Flavors & Fragrances Inc., Global Health and Nutrition Science, Danisco Sweeteners Oy, Kantvik, Finland
| |
Collapse
|
17
|
Huang L, Lv X, Ze X, Ma Z, Zhang X, He R, Fan J, Zhang M, Sun B, Wang F, Liu H. Combined probiotics attenuate chronic unpredictable mild stress-induced depressive-like and anxiety-like behaviors in rats. Front Psychiatry 2022; 13:990465. [PMID: 36159940 PMCID: PMC9490273 DOI: 10.3389/fpsyt.2022.990465] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 08/16/2022] [Indexed: 11/23/2022] Open
Abstract
Increasing evidence indicated that probiotics can be effective in improving behaviors similar to depression and anxiety disorders. However, the underlying mechanisms remain unclear, as is the effects of single vs. combined probiotics on depression and anxiety. This study aimed to determine whether combined probiotics could attenuate depressive-like and anxiety-like behavior induced by chronic unpredictable mild stress (CUMS) and its potential mechanisms. Rats underwent CUMS treatment and then administered Lactobacillus rhamnosus HN001 (HN001) or Bifidobacterium animalis subsp. lactis HN019 (HN019), alone or in combination. Levels of neurotransmitters, inflammatory factors, and the gut microbiota were measured. HN001 and (or) HN019 treatment improved depressive-like and anxiety-like behavior in rats, including increased moving distance and exploratory behavior (p < 0.05). In addition, altered gut microbiota structure induced by CUMS was amended by HN001 and/or HN019 (p < 0.05). HN001 and/or HN019 intervention also remarkably normalized levels of 5-HT, DA, NE, HVA, DOPAC, HIAA, TNF-α, IL-6, IL-18 and IL-1β in CUMS rats (p < 0.05). Furthermore, the effects of combined probiotics on decreasing inflammation and improved gut microbiota (Chao1 index and ACE index, p < 0.05) were superior to the single probiotics. Moreover, spearman analysis showed a certain correlation between the different microbiota, such as Firmicutes, Bacteroidetes, Verrucomicrobias, Proteobacterias and Actinobacterias, and inflammation and neurotransmitters. These findings suggested that CUMS induced depressive and anxiety-like behaviors can be alleviated by the combination of probiotics, which was possibly associated with the alterations in the gut microbiota composition and increased neurotransmitters and decreased inflammatory factors.
Collapse
Affiliation(s)
- Li Huang
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Center for International Collaborative Research on Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin, China
| | - Xia Lv
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Center for International Collaborative Research on Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin, China
| | - Xiaolei Ze
- BYHEALTH Institute of Nutrition & Health, Guangzhou, China
| | - Zewei Ma
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Center for International Collaborative Research on Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin, China
| | - Xuguang Zhang
- BYHEALTH Institute of Nutrition & Health, Guangzhou, China
| | - Ruikun He
- BYHEALTH Institute of Nutrition & Health, Guangzhou, China
| | - Junting Fan
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Center for International Collaborative Research on Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin, China
| | - Meilin Zhang
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Center for International Collaborative Research on Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin, China
| | - Boran Sun
- Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Center for International Collaborative Research on Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin, China.,Department of Epidemiology and Statistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Fang Wang
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Huan Liu
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Center for International Collaborative Research on Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin, China
| |
Collapse
|
18
|
Adjunctive Probiotics Alleviates Asthmatic Symptoms via Modulating the Gut Microbiome and Serum Metabolome. Microbiol Spectr 2021; 9:e0085921. [PMID: 34612663 PMCID: PMC8510161 DOI: 10.1128/spectrum.00859-21] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Asthma is a multifactorial disorder, and microbial dysbiosis enhances lung inflammation and asthma-related symptoms. Probiotics have shown anti-inflammatory effects and could regulate the gut-lung axis. Thus, a 3-month randomized, double-blind, and placebo-controlled human trial was performed to investigate the adjunctive efficacy of probiotics in managing asthma. Fifty-five asthmatic patients were randomly assigned to a probiotic group (n = 29; received Bifidobacterium lactis Probio-M8 powder and Symbicort Turbuhaler) and a placebo group (n = 26; received placebo and Symbicort Turbuhaler), and all 55 subjects provided details of their clinical history and demographic data. However, only 31 patients donated a complete set of fecal and blood samples at all three time points for further analysis. Compared with those of the placebo group, co-administering Probio-M8 with Symbicort Turbuhaler significantly decreased the fractional exhaled nitric oxide level at day 30 (P = 0.049) and improved the asthma control test score at the end of the intervention (P = 0.023). More importantly, the level of alveolar nitric oxide concentration decreased significantly among the probiotic receivers at day 30 (P = 0.038), and the symptom relief effect was even more obvious at day 90 (P = 0.001). Probiotic co-administration increased the resilience of the gut microbiome, which was reflected by only minor fluctuations in the gut microbiome diversity (P > 0.05, probiotic receivers; P < 0.05, placebo receivers). Additionally, the probiotic receivers showed significantly changes in some species-level genome bins (SGBs), namely, increases in potentially beneficial species Bifidobacterium animalis, Bifidobacterium longum, and Prevotella sp. CAG and decreases in Parabacteroides distasonis and Clostridiales bacterium (P < 0.05). Compared with that of the placebo group, the gut metabolic potential of probiotic receivers exhibited increased levels of predicted microbial bioactive metabolites (linoleoyl ethanolamide, adrenergic acid, erythronic acid) and serum metabolites (5-dodecenoic acid, tryptophan, sphingomyelin) during/after intervention. Collectively, our results suggested that co-administering Probio-M8 synergized with conventional therapy to alleviate diseases associated with the gut-lung axis, like asthma, possibly via activating multiple anti-inflammatory pathways. IMPORTANCE The human gut microbiota has a potential effect on the pathogenesis of asthma and is closely related to the disease phenotype. Our trial has demonstrated that co-administering Probio-M8 synergized with conventional therapy to alleviate asthma symptoms. The findings of the present study provide new insights into the pathogenesis and treatment of asthma, mechanisms of novel therapeutic strategies, and application of probiotics-based therapy.
Collapse
|
19
|
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: 36] [Impact Index Per Article: 9.0] [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
|
20
|
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
|
21
|
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: 37] [Impact Index Per Article: 9.3] [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
|
22
|
Huidrom S. Therapeutic Approach of Probiotics in Children with Atopic Dermatitis. Antiinflamm Antiallergy Agents Med Chem 2021; 20:2-9. [PMID: 31899681 DOI: 10.2174/1871523019666200102110317] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/16/2019] [Accepted: 11/28/2019] [Indexed: 12/13/2022]
Abstract
Pediatric atopic dermatitis (AD) is a chronic, relapsing inflammatory skin disease, affecting 20% of children all over the world especially in developed countries. The global prevalence of AD in children has been increasing over recent years. This chronic inflammatory skin disease causes economic and social burden to the family. The exact cause of AD is not known, however recent studies suggest that the imbalance of microflora present in the gut leads to AD. The current treatment of AD involves the application of moisturizer, topical corticosteroids, antihistamines and antibiotics. This line of treatment of AD in children has many side effects. An alternative novel therapeutic approach has to be explored to combat this chronic skin disease. In recent years, there has been increasing interest in the use of probiotics in the modulation of gut microbiota for the management of AD. Many research studies showed that the administration of probiotics gives positive results in the prevention and treatment of AD in children, however, the results are not consistent and conclusive. In this review, the phenomenon that the dysbiosis of the gut flora contributes to the development of AD is addressed and clinical evidence of probiotics in the prevention and treatment of AD children is also summarised.
Collapse
Affiliation(s)
- Sangeeta Huidrom
- Department of Pharmacology, Shri Guru Ram Rai Institute of Medical and Health Sciences, Dehradun-248001, India
| |
Collapse
|
23
|
Gan W, Yang F, Meng J, Liu F, Liu S, Xian J. Comparing the nasal bacterial microbiome diversity of allergic rhinitis, chronic rhinosinusitis and control subjects. Eur Arch Otorhinolaryngol 2021; 278:711-718. [PMID: 32860131 DOI: 10.1007/s00405-020-06311-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 08/19/2020] [Indexed: 12/18/2022]
Abstract
PURPOSE Until now, the microbiome of the nasal cavity and its contribution to nasal mucosal disease has remained poorly understood. The advent of cultivation-free molecular methods makes it possible to characterize the total microbiome of the nasal cavity. We sought to assess the microbial diversity and composition of the middle meatus in allergic rhinitis (AR) patients, chronic rhinosinusitis patients without polyps (CRSsNP) and a control population to determine the microbiota associated with the pathogenesis of AR and CRSsNP. METHODS Microbial characterization was determined using 16S rRNA gene sequencing of 122 nasal swabs collected from patients with AR (n = 52) and CRSsNP (n = 37), and from healthy controls (n = 33). RESULTS There was no difference in nasal microbiome richness and diversity among the three groups, and the dominant phyla were similar among three groups including Firmicutes, Proteobacteria, Actinobacteria and Bacteroidetes. However, Spirochaetae abundance was significantly higher in AR than in the control group after FDR correction (FDR p = 0.021). At the genus level, although there was no statistical significance after FDR correction, there was a trend that Pseudomonas and Peptostreptococcaceae abundance were higher in AR than in controls (p = 0.005, p = 0.005) and CRSsNP (p = 0.023, p = 0.034); Lactobacillus abundance was lower in AR than in controls (p = 0.021); Moraxella abundance was lower in CRSsNP than in controls (p = 0.006); Haemophilus abundance was higher in CRSsNP than in AR (p = 0.003) but lower in AR than in controls (p = 0.018). CONCLUSION These results suggested that microbial dysbiosis may play a role in the pathogenesis of heterogeneous nasal mucosal inflammation.
Collapse
Affiliation(s)
- Weigang Gan
- Department of Otorhinolaryngology-Head and Neck Surgery, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Fengjuan Yang
- Department of Otorhinolaryngology, Nanchong Fifth People's Hospital, Nanchong, Sichuan, People's Republic of China
| | - Juan Meng
- Department of Otorhinolaryngology-Head and Neck Surgery, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| | - Feng Liu
- Department of Otorhinolaryngology-Head and Neck Surgery, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Shixi Liu
- Department of Otorhinolaryngology-Head and Neck Surgery, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Junming Xian
- Department of Otorhinolaryngology-Head and Neck Surgery, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, People's Republic of China
| |
Collapse
|
24
|
Zommiti M, Feuilloley MGJ, Connil N. Update of Probiotics in Human World: A Nonstop Source of Benefactions till the End of Time. Microorganisms 2020; 8:E1907. [PMID: 33266303 PMCID: PMC7760123 DOI: 10.3390/microorganisms8121907] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/23/2020] [Accepted: 11/23/2020] [Indexed: 02/06/2023] Open
Abstract
Lactic acid bacteria (LAB) are known for their biotechnological potential. Moreover, LAB are distinguished by amazing criteria: Adjusting the intestinal environment, inhibiting pathogenic microbes in the gastrointestinal tract, ability to reduce pathogen adhesion activity, improving the balance of the microbiota inside the intestine, capabilities of regulating intestinal mucosal immunity, and maintaining intestinal barrier function. The escalating number of research and studies about beneficial microorganisms and their impact on promoting health has attracted a big interest in the last decades. Since antiquity, various based fermented products of different kinds have been utilized as potential probiotic products. Nevertheless, the current upsurge in consumers' interest in bioalternatives has opened new horizons for the probiotic field in terms of research and development. The present review aims at shedding light on the world of probiotics, a continuous story of astonishing success in various fields, in particular, the biomedical sector and pharmaceutical industry, as well as to display the importance of probiotics and their therapeutic potential in purpose to compete for sturdy pathogens and to struggle against diseases and acute infections. Shadows and future trends of probiotics use are also discussed.
Collapse
Affiliation(s)
- Mohamed Zommiti
- Unité de Protéomique Fonctionnelle et Potentiel Nutraceutique de la Biodiversité de Tunisie, Institut Supérieur des Sciences Biologiques Appliquées de Tunis, Université Tunis El-Manar, Tunis 1006, Tunisia
| | - Marc G. J. Feuilloley
- Laboratoire de Microbiologie Signaux et Microenvironnement (LMSM) EA 4312, Université de Rouen Normandie, Normandie Université, F-27000 Evreux, France; (M.G.J.F.); (N.C.)
| | - Nathalie Connil
- Laboratoire de Microbiologie Signaux et Microenvironnement (LMSM) EA 4312, Université de Rouen Normandie, Normandie Université, F-27000 Evreux, France; (M.G.J.F.); (N.C.)
| |
Collapse
|
25
|
Peñalver Bernabé B, Maki PM, Dowty SM, Salas M, Cralle L, Shah Z, Gilbert JA. Precision medicine in perinatal depression in light of the human microbiome. Psychopharmacology (Berl) 2020; 237:915-941. [PMID: 32065252 DOI: 10.1007/s00213-019-05436-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 12/11/2019] [Indexed: 12/17/2022]
Abstract
Perinatal depression is the most common complication of pregnancy and affects the mother, fetus, and infant. Recent preclinical studies and a limited number of clinical studies have suggested an influence of the gut microbiome on the onset and course of mental health disorders. In this review, we examine the current state of knowledge regarding genetics, epigenetics, heritability, and neuro-immuno-endocrine systems biology in perinatal mood disorders, with a particular focus on the interaction between these factors and the gut microbiome, which is mediated via the gut-brain axis. We also provide an overview of experimental and analytical methods that are currently available to researchers interested in elucidating the influence of the gut microbiome on mental health disorders during pregnancy and postpartum.
Collapse
Affiliation(s)
- Beatriz Peñalver Bernabé
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois, United States.
| | - Pauline M Maki
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
- Department of Psychology, University of Illinois at Chicago, Chicago, IL, USA
- Department of Obstetrics and Gynecology, University of Illinois at Chicago, Chicago, IL, USA
| | - Shannon M Dowty
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | - Mariana Salas
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Lauren Cralle
- University of Massachusetts Medical School, Worcester, MA, USA
| | - Zainab Shah
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | - Jack A Gilbert
- Scripts Oceanographic Institute, University of California San Diego, La Jolla, CA, USA
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
26
|
Bawany F, Beck LA, Järvinen KM. Halting the March: Primary Prevention of Atopic Dermatitis and Food Allergies. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2020; 8:860-875. [PMID: 32147139 PMCID: PMC7355223 DOI: 10.1016/j.jaip.2019.12.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 12/13/2022]
Abstract
Atopic dermatitis (AD) is one of the most common inflammatory skin conditions, affecting 15% to 30% of children and 2% to 10% of adults. Population-based studies suggest that having AD is associated with subsequent development of other atopic diseases, in what is known as the "atopic march." We will provide an overview of studies that investigate primary prevention strategies for the first 2 diseases in the march, namely, AD and food allergies (FA). These strategies include emollients, breastfeeding, microbial exposures, probiotics, vitamin D and UV light, water hardness, and immunotherapy. Some studies, including randomized controlled trials on emollients and microbial supplementation, have found encouraging results; however, the evidence remains limited and contradictory. With regard to breastfeeding, microbial and lifestyle exposures, vitamin D and UV light, water hardness, and immunotherapy, the lack of randomized controlled trials makes it difficult to draw definitive conclusions. Current American Academy of Pediatrics guidelines support the idea that breastfeeding for 3 to 4 months can decrease AD incidence in children less than 2 years old. Recommendations regarding a direct relationship between breastfeeding on FA, however, cannot be made because of insufficient data. Regarding microbial supplementation, most guidelines do not recommend probiotics or prebiotics for the purpose of preventing allergic diseases because of limited evidence. Before definitive conclusions can be made regarding these interventions, more well-designed, longitudinal, and randomized controlled trials, particularly in at-risk populations, are required.
Collapse
Affiliation(s)
- Fatima Bawany
- School of Medicine and Dentistry, University of Rochester Medical Center, Rochester, NY
| | - Lisa A Beck
- Department of Dermatology, University of Rochester Medical Center, Rochester, NY.
| | - Kirsi M Järvinen
- Department of Pediatrics, Division of Allergy and Immunology & Center for Food Allergy, University of Rochester Medical Center, Rochester, NY
| |
Collapse
|
27
|
Lactobacillus fermentum CECT5716 Supplementation in Rats during Pregnancy and Lactation Impacts Maternal and Offspring Lipid Profile, Immune System and Microbiota. Cells 2020; 9:cells9030575. [PMID: 32121244 PMCID: PMC7140451 DOI: 10.3390/cells9030575] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 12/12/2022] Open
Abstract
Probiotics have shown potential for their use in early life. This study aimed to investigate whether the administration of Lactobacillus fermentum CECT5716 during pregnancy and lactation periods impacts maternal and offspring plasma lipid profile, immune system and microbiota. Rats were supplemented with the probiotic during gestation and two weeks of lactation. After supplementation, although the microbiota composition was not affected, the probiotic strain was detected in all cecal contents of dams and in some of their pups. Dams showed reduced proportion of T cytotoxic cells in the mesenteric lymph nodes, modulation of intestinal cytokines (IL-10 and IL-12) and changes in plasma fatty acids (20:0, 22:0, 20:5 n-3, and 18:3 n-6). Pups showed changes in immunoglobulins (intestinal IgA and plasmatic IgG2a and IgG2c) and fatty acid profile (17:0, 22:0, and 18:2 n-6). Overall, Lactobacillus fermentum CECT5716 supplementation contributed to beneficially modulating the immune system of the mother and its offspring.
Collapse
|
28
|
Szari S, Quinn JA. Supporting a Healthy Microbiome for the Primary Prevention of Eczema. Clin Rev Allergy Immunol 2020; 57:286-293. [PMID: 31309394 DOI: 10.1007/s12016-019-08758-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Eczema is increasing worldwide with associated increases in health costs and decreases in quality of life. There are many factors that are speculated to interact in the development of eczema including genetics and environmental exposures. Prevention of the development of eczema may prevent the further development of food allergies and asthma. This concept has prompted a variety of research into the area of primary prevention of eczema in infants. This exploration includes a growing body of research examining infants supplemented with probiotics, prebiotics, or both (synbiotics) often compared with their breastfed counterparts. The goal of this paper is to examine the evidence for manipulating the microbiome in the prevention of eczema. Several strains of probiotics, compositions of prebiotics, and varied combinations of both are commercially available. Evidence supports altering the microbiome in infants at high risk of atopy who are not able to breastfeed with Lactobacillus strains when given both prenatally followed by prolonged use (greater than 6 months) postnatally for the primary prevention of eczema. Prebiotics have also been shown beneficial for primary prevention of eczema in formula-fed infants with prolonged use greater than 6 months. These findings are in keeping with the World Allergy Organization (WAO) recommendations that support interventions to manipulate the microbiome with both probiotics and prebiotics.
Collapse
Affiliation(s)
- Sofia Szari
- Department of Allergy-Immunology, Wilford Hall Ambulatory Surgical Center, Lackland Air Force Base, San Antonio, TX, USA.
| | - James A Quinn
- Department of Allergy-Immunology, Wilford Hall Ambulatory Surgical Center, Lackland Air Force Base, San Antonio, TX, USA
| |
Collapse
|
29
|
Peldan P, Kukkonen A, Savilahti E, Kuitunen M. Perinatal Probiotic Mixture and Development of Allergic Sensitization up to 13 Years of Age. Int Arch Allergy Immunol 2020; 181:270-277. [DOI: 10.1159/000504915] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 11/20/2019] [Indexed: 11/19/2022] Open
|
30
|
Sestito S, D'Auria E, Baldassarre ME, Salvatore S, Tallarico V, Stefanelli E, Tarsitano F, Concolino D, Pensabene L. The Role of Prebiotics and Probiotics in Prevention of Allergic Diseases in Infants. Front Pediatr 2020; 8:583946. [PMID: 33415087 PMCID: PMC7783417 DOI: 10.3389/fped.2020.583946] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 11/23/2020] [Indexed: 12/15/2022] Open
Abstract
Allergic diseases have been linked to genetic and/or environmental factors, such as antibiotic use, westernized high fat and low fiber diet, which lead to early intestinal dysbiosis, and account for the rise in allergy prevalence, especially in western countries. Allergic diseases have shown reduced microbial diversity, including fewer lactobacilli and bifidobacteria, within the neonatal microbiota, before the onset of atopic diseases. Raised interest in microbiota manipulating strategies to restore the microbial balance for atopic disease prevention, through prebiotics, probiotics, or synbiotics supplementation, has been reported. We reviewed and discussed the role of prebiotics and/or probiotics supplementation for allergy prevention in infants. We searched PubMed and the Cochrane Database using keywords relating to "allergy" OR "allergic disorders," "prevention" AND "prebiotics" OR "probiotics" OR "synbiotics." We limited our evaluation to papers of English language including children aged 0-2 years old. Different products or strains used, different period of intervention, duration of supplementation, has hampered the draw of definitive conclusions on the clinical impact of probiotics and/or prebiotics for prevention of allergic diseases in infants, except for atopic dermatitis in infants at high-risk. This preventive effect on eczema in high-risk infants is supported by clear evidence for probiotics but only moderate evidence for prebiotic supplementation. However, the optimal prebiotic or strain of probiotic, dose, duration, and timing of intervention remains uncertain. Particularly, a combined pre- and post-natal intervention appeared of stronger benefit, although the definition of the optimal intervention starting time during gestation, the timing, and duration in the post-natal period, as well as the best target population, are still an unmet need.
Collapse
Affiliation(s)
- Simona Sestito
- Pediatric Unit, Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Enza D'Auria
- Department of Pediatrics, Vittore Buzzi Children's Hospital-University of Milan, Milan, Italy
| | - Maria Elisabetta Baldassarre
- Neonatology and Neonatal Intensive Care Unit, Department of Biomedical Science and Human Oncology, "Aldo Moro" University of Bari, Bari, Italy
| | - Silvia Salvatore
- Department of Pediatrics, Ospedale "F. Del Ponte", University of Insubria, Varese, Italy
| | - Valeria Tallarico
- Pediatric Unit, Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Ettore Stefanelli
- Pediatric Unit, Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Flora Tarsitano
- Pediatric Unit, Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Daniela Concolino
- Pediatric Unit, Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.,Department of Health Sciences, School of Medicine and Surgery, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Licia Pensabene
- Pediatric Unit, Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| |
Collapse
|
31
|
Crane J, Barthow C, Kang J, Hood F, Stanley T, Wickens K. Probiotics for humans: hoax, hype, hope, or help. J R Soc N Z 2019. [DOI: 10.1080/03036758.2019.1692364] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Julian Crane
- Dept. of Medicine, University of Otago, Wellington, New Zealand
| | | | - Janice Kang
- Dept. of Medicine, University of Otago, Wellington, New Zealand
| | - Fiona Hood
- Dept. of Medicine, University of Otago, Wellington, New Zealand
| | - Thorsten Stanley
- Dept. of Paediatrics, University of Otago, Wellington, New Zealand
| | - Kristin Wickens
- Dept. of Medicine, University of Otago, Wellington, New Zealand
| |
Collapse
|
32
|
Amalia N, Orchard D, Francis KL, King E. Systematic review and meta‐analysis on the use of probiotic supplementation in pregnant mother, breastfeeding mother and infant for the prevention of atopic dermatitis in children. Australas J Dermatol 2019; 61:e158-e173. [DOI: 10.1111/ajd.13186] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 08/31/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Nasya Amalia
- Faculty of Medicine Universitas Indonesia Jakarta Indonesia
- Faculty of Medical Science University of Melbourne Melbourne Australia
| | - David Orchard
- Faculty of Medical Science University of Melbourne Melbourne Australia
- Dermatology DepartmentThe Royal Children's Hospital Melbourne Australia
| | - Kate Louise Francis
- Clinical Epidemiology and Biostatistics Unit Murdoch Children's Research Institute The Royal Children's Hospital Melbourne Australia
| | - Emma King
- Faculty of Medical Science University of Melbourne Melbourne Australia
- Dermatology DepartmentThe Royal Children's Hospital Melbourne Australia
| |
Collapse
|
33
|
Kleniewska P, Pawliczak R. The influence of apocynin, lipoic acid and probiotics on antioxidant enzyme levels in the pulmonary tissues of obese asthmatic mice. Life Sci 2019; 234:116780. [PMID: 31430453 DOI: 10.1016/j.lfs.2019.116780] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 08/07/2019] [Accepted: 08/16/2019] [Indexed: 12/25/2022]
Abstract
Bronchial asthma and obesity are common health problems. Obesity is already responsible for 300,000 deaths per year. AIMS The aim of the present study was to assess whether apocynin, alpha lipoic acid and probiotic administration in combination with low-fat diet supplementation influences the levels of antioxidant enzymes in the pulmonary tissues of obese asthmatic mice. MAIN METHODS The study was performed on male C57/BL6 mice divided into 10 groups: (I) control; (II) asthma; (III) obesity; (IV) asthma + obesity; (V) asthma + obesity + apocynin p.o. 15 mg/kg/day for 12 weeks; (VI) asthma + obesity + low-fat diet for 12 weeks; (VII) asthma + obesity + low-fat diet for 12 weeks with apocynin p.o. 15 mg/kg/day; (VIII) asthma + obesity + low-fat diet with probiotics for 12 weeks; (IX) asthma + obesity + low-fat diet for 12 weeks with lipoic acid p.o. 100 mg/kg/day for 12 weeks; (X) asthma + obesity + standard diet with probiotics for 12 weeks. Superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx) and glutathione reductase (GR) activity were examined. The administration of apocynin alone and apocynin in combination with a low-fat diet resulted in a significant increase in SOD values (respectively p < 0.001; p = 0.010). Application of probiotics resulted in a decrease in CAT activity (p = 0.037) and an increase in GPx activity (p < 0.001) compared to obese asthmatic mice. The administration of lipoic acid resulted in an increase in GR activity (p = 0.024 vs. control). KEY FINDINGS Supplementation containing apocynin, lipoic acid and probiotics has a positive influence on the antioxidant capacity of the pulmonary tissues of obese asthmatic mice. SIGNIFICANCE These results may contribute to the development of new therapeutic approaches.
Collapse
Affiliation(s)
- Paulina Kleniewska
- Department of Immunopathology, Faculty of Biomedical Sciences and Postgraduate Training, Medical University of Lodz, ul. Zeligowskiego 7/9, 90-752 Lodz, Poland.
| | - Rafał Pawliczak
- Department of Immunopathology, Faculty of Biomedical Sciences and Postgraduate Training, Medical University of Lodz, ul. Zeligowskiego 7/9, 90-752 Lodz, Poland
| |
Collapse
|
34
|
Beeinflussung des Mikrobioms durch Probiotika in der pädiatrischen Praxis. Monatsschr Kinderheilkd 2019. [DOI: 10.1007/s00112-019-0690-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
35
|
Murphy R, Morgan X, Wang X, Wickens K, Purdie G, Fitzharris P, Otal A, Lawley B, Stanley T, Barthow C, Crane J, Mitchell E, Tannock G. Eczema-protective probiotic alters infant gut microbiome functional capacity but not composition: sub-sample analysis from a RCT. Benef Microbes 2019; 10:5-17. [DOI: 10.3920/bm2017.0191] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Probiotic Lactobacillus rhamnosus HN001 given in early life has been shown to reduce infant eczema risk, but its effect on gut microbiota development has not been quantitatively and functionally examined. The aim of this study was to investigate the impact of early life probiotic exposure on the composition and functional capacity of infant gut microbiota from birth to 2 years considering the effects of age, delivery mode, antibiotics, pets and eczema. We performed shotgun metagenomic sequencing analysis of 650 infant faecal samples, collected at birth, 3, 12, and 24 months, as part of a randomised, controlled, 3-arm trial assessing the effect of L. rhamnosus HN001, Bifidobacterium animalis subsp. lactis HN019 supplementation on eczema development in 474 infants. There was a 50% reduced eczema risk in the HN001 probiotic group compared to placebo. Both mothers (from 35 weeks gestation until 6 months post-partum if breastfeeding) and infants (from birth to 2 years) received either a placebo or one of two probiotics, L. rhamnosus HN001 (6×109 cfu), or B. animalis subsp. lactis HN019 (9×109 cfu). L. rhamnosus HN001 probiotic supplementation was associated with increased overall glycerol-3 phosphate transport capacity and enrichment of L. rhamnosus. There were no other significant changes in infant gut microbiota composition or diversity. Increased capacity to transport glycerol-3-phosphate was positively correlated with relative abundance of L. rhamnosus. Children who developed eczema had gut microbiota with increased capacity for glycosaminoglycan degradation and flagellum assembly but had no significant differences in microbiota composition or diversity. Early life HN001 probiotic use is associated with both increased L. rhamnosus and increased infant gut microbiota functional capacity to transport glycerol-3 phosphate. The mechanistic relationship of such functional alteration in gut microbiota with reduced eczema risk and long-term health merits further investigation.
Collapse
Affiliation(s)
- R. Murphy
- Faculty of Medical and Health Sciences, University of Auckland, Park Road, Grafton, 1142 Auckland, New Zealand
| | - X.C. Morgan
- Department of Microbiology and Immunology, University of Otago, 720 Cumberland St, Dunedin 9016 New Zealand
| | - X.Y. Wang
- Department of Microbiology and Immunology, University of Otago, 720 Cumberland St, Dunedin 9016 New Zealand
| | - K. Wickens
- Department of Medicine, University of Otago, P.O. Box 7343, Wellington South, 6021 Wellington, New Zealand
| | - G. Purdie
- Department of Medicine, University of Otago, P.O. Box 7343, Wellington South, 6021 Wellington, New Zealand
| | - P. Fitzharris
- Department of Immunology, Auckland City Hospital, 2 Park Road, Grafton, 1023 Auckland, New Zealand
| | - A. Otal
- Department of Microbiology and Immunology, University of Otago, 720 Cumberland St, Dunedin 9016 New Zealand
| | - B. Lawley
- Department of Microbiology and Immunology, University of Otago, 720 Cumberland St, Dunedin 9016 New Zealand
| | - T. Stanley
- Department of Medicine, University of Otago, P.O. Box 7343, Wellington South, 6021 Wellington, New Zealand
| | - C. Barthow
- Department of Medicine, University of Otago, P.O. Box 7343, Wellington South, 6021 Wellington, New Zealand
| | - J. Crane
- Department of Medicine, University of Otago, P.O. Box 7343, Wellington South, 6021 Wellington, New Zealand
| | - E.A. Mitchell
- Department of Paediatrics, University of Auckland, Auckland 1142, New Zealand
| | - G.W. Tannock
- Microbiome Otago, University of Otago, 720 Cumberland St., 9016 Dunedin, New Zealand
- Department of Microbiology and Immunology, University of Otago, 720 Cumberland St, Dunedin 9016 New Zealand
| |
Collapse
|
36
|
Wei X, Jiang P, Liu J, Sun R, Zhu L. Association between probiotic supplementation and asthma incidence in infants: a meta-analysis of randomized controlled trials. J Asthma 2019; 57:167-178. [PMID: 30656984 DOI: 10.1080/02770903.2018.1561893] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Objective: The increased social and economic burdens for asthma in infants make the prevention of asthma a major public health goal. Probiotics may reduce the risk of asthma in infants. However, randomized controlled trials (RCTs) have shown mixed efficacy outcomes. We performed a meta-analysis of RCTs to investigate whether probiotics are associated with a lower asthma incidence in infants. Methods: The PubMed, Cochrane library, and EMBASE databases were systematically searched from the inception dates to August 2018. RCTs comparing the effects of probiotic supplements with a placebo for asthma or wheeze incidence in infants were included. A meta-analysis was performed to calculate risk ratio (RR) and 95% confidence interval (CI) using the Mantel-Haenszel statistical method. Results: A total of 19 randomized trials involving 5157 children fulfilled the inclusion criteria. There was no significant association of probiotics with risk of asthma (RR, 0.94 [95% CI, 0.82-1.09]) or wheeze (RR, 0.97 [95% CI, 0.88-1.06]) compared with placebo. Subgroup analysis by asthma risk showed that probiotics significantly reduced wheeze incidence among infants with atopy disease (RR, 0.61 [95% CI, 0.42-0.90]), but no significant associations were found in the other subgroup analyses by participants receiving the intervention, timing of intervention, prevention regimen, probiotic organism, duration of intervention, and duration of follow-up. Conclusions: The use of probiotic supplementation compared with placebo was not associated with a lower risk of asthma in infants. These findings do not support recommendation to use probiotics in the prevention of asthma in infants.
Collapse
Affiliation(s)
- Xiaochen Wei
- Department of Pharmacy, Tianjin First Central Hospital, Tianjin, People's Republic of China
| | - Ping Jiang
- Department of Respiration, Tianjin First Central Hospital, Tianjin, People's Republic of China
| | - Jiangbo Liu
- Department of Respiration, Tianjin First Central Hospital, Tianjin, People's Republic of China
| | - Rongfei Sun
- Department of Respiration, Tianjin First Central Hospital, Tianjin, People's Republic of China
| | - Liqin Zhu
- Department of Pharmacy, Tianjin First Central Hospital, Tianjin, People's Republic of China
| |
Collapse
|
37
|
Li X, Peng Y, Li Z, Christensen B, Heckmann AB, Stenlund H, Lönnerdal B, Hernell O. Feeding Infants Formula With Probiotics or Milk Fat Globule Membrane: A Double-Blind, Randomized Controlled Trial. Front Pediatr 2019; 7:347. [PMID: 31552203 PMCID: PMC6736587 DOI: 10.3389/fped.2019.00347] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 08/05/2019] [Indexed: 12/16/2022] Open
Abstract
Purpose: To evaluate effects on growth and infection rates of supplementing infant formula with the probiotic Lactobacillus paracasei ssp. paracasei strain F19 (F19) or bovine milk fat globule membrane (MFGM). Methods: In a double-blind, randomized controlled trial, 600 infants were randomized to a formula supplemented with F19 or MFGM, or to standard formula (SF). A breastfed group was recruited as reference (n = 200).The intervention lasted from age 21 ± 7 days until 4 months, and infants were followed until age one year. Results: Both experimental formulas were well tolerated and resulted in high compliance. The few reported adverse events were not likely related to formula, with the highest rates in the SF group, significantly higher than for the F19-supplemented infants (p = 0.046). Weight or length gain did not differ during or after the intervention among the formula-fed groups, with satisfactory growth. During the intervention, overall, the experimental formula groups did not have more episodes of diarrhea, fever, or days with fever than the breastfed infants. However, compared to the breastfed infants, the SF group had more fever episodes (p = 0.021) and days with fever (p = 0.036), but not diarrhea. Compared with the breastfed group, the F19-supplemented infants but not the other two formula groups had more visits/unscheduled hospitalizations (p = 0.015) and borderline more episodes of upper respiratory tract infections (p = 0.048). Conclusions: Both the MFGM- and F19-supplemented formulas were safe and well-tolerated, leading to few adverse effects, similar to the breastfed group and unlike the SF group. During the intervention, the MFGM-supplemented infants did not differ from the breastfed infants in any primary outcome.
Collapse
Affiliation(s)
- Xiaonan Li
- Department of Children Health Care, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yongmei Peng
- Department of Children Health Care, Children's Hospital of Fudan University, Shanghai, China
| | - Zailing Li
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| | | | | | - Hans Stenlund
- Epidemiology and Global Health, Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Bo Lönnerdal
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Olle Hernell
- Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, Sweden
| |
Collapse
|
38
|
Kallio S, Kukkonen AK, Savilahti E, Kuitunen M. Perinatal probiotic intervention prevented allergic disease in a Caesarean‐delivered subgroup at 13‐year follow‐up. Clin Exp Allergy 2018; 49:506-515. [DOI: 10.1111/cea.13321] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 10/12/2018] [Accepted: 10/15/2018] [Indexed: 02/01/2023]
Affiliation(s)
- Sampo Kallio
- Children's Hospital University of Helsinki and Helsinki University Hospital Helsinki Finland
| | - Anna Kaarina Kukkonen
- Skin and Allergy Hospital University of Helsinki and Helsinki University Hospital Helsinki Finland
| | - Erkki Savilahti
- Children's Hospital University of Helsinki and Helsinki University Hospital Helsinki Finland
| | - Mikael Kuitunen
- Children's Hospital University of Helsinki and Helsinki University Hospital Helsinki Finland
| |
Collapse
|
39
|
Grev J, Berg M, Soll R, Cochrane Neonatal Group. Maternal probiotic supplementation for prevention of morbidity and mortality in preterm infants. Cochrane Database Syst Rev 2018; 12:CD012519. [PMID: 30548483 PMCID: PMC6516999 DOI: 10.1002/14651858.cd012519.pub2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND Inflammation may contribute to preterm birth and to morbidity of preterm infants. Preterm infants are at risk for alterations in the normal protective microbiome. Oral probiotics administered directly to preterm infants have been shown to decrease the risk for severe necrotizing enterocolitis (NEC) as well as the risk of death, but there are safety concerns about administration of probiotics directly to preterm infants. Through decreasing maternal inflammation, probiotics may play a role in preventing preterm birth and/or decreasing the inflammatory milieu surrounding delivery of preterm infants, and may alter the microbiome of the preterm infant when given to mothers during pregnancy. Probiotics given to mothers after birth of preterm infants may effect infant bacterial colonization, which could potentially reduce the incidence of NEC. OBJECTIVES 1. To compare the efficacy of maternal probiotic administration versus placebo or no intervention in mothers during pregnancy for the prevention of preterm birth and the prevention of morbidity and mortality of infants born preterm.2. To compare the efficacy of maternal probiotic administration versus placebo, no intervention, or neonatal probiotic administration in mothers of preterm infants after birth on the prevention of mortality and preterm infant morbidities such as NEC. SEARCH METHODS We used the standard search strategy of Cochrane Neonatal to search the Cochrane Central Register of Controlled Trials (CENTRAL 2017, Issue 2), MEDLINE via PubMed (1966 to 21 March 2017), Embase (1980 to 21 March 2017), and CINAHL (1982 to 21 March 2017). We also searched clinical trials databases, conference proceedings, and the reference lists of retrieved articles for randomized controlled trials and quasi-randomized trials. SELECTION CRITERIA We included randomized controlled trials in the review if they administered oral probiotics to pregnant mothers at risk for preterm birth, or to mothers of preterm infants after birth. Quasi-randomized trials were eligible for inclusion, but none were identified. Studies enrolling pregnant women needed to administer probiotics at < 36 weeks' gestation until the trimester of birth. Probiotics considered were of the genera Lactobacillus, Bifidobacterium or Saccharomyces. DATA COLLECTION AND ANALYSIS We used the standard methods of the Cochrane Collaboration and Cochrane Neonatal to determine the methodologic quality of studies, and for data collection and analysis. MAIN RESULTS We included 12 eligible trials with a total of 1450 mothers and 1204 known infants. Eleven trials administered probiotics to mothers during pregnancy and one trial administered probiotics to mothers after birth of their preterm infants. No studies compared maternal probiotic administration directly with neonatal administration. Included prenatal trials were highly variable in the indication for the trial, the gestational age and duration of administration of probiotics, as well as the dose and formulation of the probiotics. The pregnant women included in these trials were overall at low risk for preterm birth. In a meta-analysis of trial data, oral probiotic administration to pregnant women did not reduce the incidence of preterm birth < 37 weeks (typical risk ratio (RR) 0.92, 95% confidence interval (CI) 0.32 to 2.67; 4 studies, 518 mothers and 506 infants), < 34 weeks (typical risk difference (RD) 0.00, 95% CI -0.02 to 0.02; 2 studies, 287 mothers and infants), the incidence of infant mortality (typical RD 0.00, 95% CI -0.02 to 0.02; 2 studies, 309 mothers and 298 infants), or the gestational age at birth (mean difference (MD) 0.15, 95% CI -0.33 to 0.63; 2 studies, 209 mothers with 207 infants).One trial studied administration of probiotics to mothers after preterm birth and included 49 mothers and 58 infants. There were no significant differences in the risk of any NEC (RR 0.44, 95% CI 0.13 to 1.46; 1 study, 58 infants), surgery for NEC (RR 0.15, 95% CI 0.01 to 2.58; 1 study, 58 infants), death (RR 0.66, 95% CI 0.06 to 6.88; 1 study, 58 infants), and death or NEC (RR 0.53, 95% CI 0.19 to 1.49; 1 study, 58 infants). There was an improvement in time to reach 50% enteral feeds in infants whose mothers received probiotics, but the estimate is imprecise (MD -9.60 days, 95% CI -19.04 to -0.16 days; 58 infants). No other improvement in any neonatal outcomes were reported. The estimates were imprecise and do not exclude the possibility of meaningful harms or benefits from maternal probiotic administration. There were no cases of culture-proven sepsis with the probiotic organism. The GRADE quality of evidence was judged to be low to very low due to inconsistency and imprecision. AUTHORS' CONCLUSIONS There is insufficient evidence to conclude whether there is appreciable benefit or harm to neonates of either oral supplementation of probiotics administered to pregnant women at low risk for preterm birth or oral supplementation of probiotics to mothers of preterm infants after birth. Oral supplementation of probiotics to mothers of preterm infants after birth may decrease time to 50% enteral feeds, however, this estimate is extremely imprecise. More research is needed for post-natal administration of probiotics to mothers of preterm infants, as well as to pregnant mothers at high risk for preterm birth.
Collapse
Affiliation(s)
| | - Marie Berg
- Johns Hopkins All Children's HospitalPediatrics/Neonatology601 Fifth Street South, Suite 501St. PetersburgFloridaUSA33606
| | - Roger Soll
- Larner College of Medicine at the University of VermontDivision of Neonatal‐Perinatal Medicine, Department of Pediatrics111 Colchester AvenueBurlingtonVermontUSA05401
| | | |
Collapse
|
40
|
Eigenmann P. Editorial comments on this issue of the Journal. Pediatr Allergy Immunol 2018; 29:787-788. [PMID: 30548707 DOI: 10.1111/pai.13003] [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: 11/01/2018] [Accepted: 11/06/2018] [Indexed: 11/29/2022]
Affiliation(s)
- Philippe Eigenmann
- Department of Pediatrics, University Hospital of Geneva, Geneva, Switzerland
| |
Collapse
|
41
|
Wickens K, Barthow C, Mitchell EA, Kang J, van Zyl N, Purdie G, Stanley T, Fitzharris P, Murphy R, Crane J. Effects of Lactobacillus rhamnosus HN001 in early life on the cumulative prevalence of allergic disease to 11 years. Pediatr Allergy Immunol 2018; 29:808-814. [PMID: 30430649 DOI: 10.1111/pai.12982] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 08/02/2018] [Accepted: 08/09/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND In a two-centre randomized placebo-controlled trial of Lactobacillus rhamnosus HN001 (HN001) (6 × 109 colony-forming units [cfu]) or Bifidobacterium lactis HN019 (HN019) (9 × 109 cfu) taken daily from 35-week gestation to 6 months' post-partum in mothers while breastfeeding and from birth to age 2 years in infants, we showed that HN001 significantly protected against eczema development at 2, 4 and 6 years and atopic sensitization at 6 years. There was no effect of HN019. We report here the findings for 11 year outcomes. METHODS At age 11 years, eczema was defined as previously using the UK Working Party's Diagnostic Criteria. Asthma, wheeze, hay fever and rhinitis were defined based on the International Study of Asthma and Allergies in Childhood (ISAAC) questions. Atopic sensitization was defined as one or more positive responses (mean wheal diameter ≥3 mm) to a panel of food and aeroallergens. Analysis was intention-to-treat using hazard ratios to assess probiotic effects on the 11-year lifetime prevalence and relative risks for point or 12-month prevalence at 11 years. RESULTS Early childhood HN001 supplementation was associated with significant reductions in the 12-month prevalence of eczema at age 11 years (relative risk [RR] = 0.46, 95% CI 0.25-0.86, P = 0.015) and hay fever (RR = 0.73, 95% CI 0.53-1.00, P = 0.047). For the lifetime prevalence, HN001 was associated with a significant reduction in atopic sensitization (hazard ratio [HR] = 0.71, 95% CI 0.51-1.00, P = 0.048), eczema (HR = 0.58, 95% CI 0.41-0.82, P = 0.002) and wheeze (HR = 0.76, 95% CI 0.57-0.99, P = 0.046). HN019 had no significant effect on these outcomes. CONCLUSION This is the first early probiotic intervention to show positive outcomes for at least the first decade of life across the spectrum of allergic disease.
Collapse
Affiliation(s)
- Kristin Wickens
- Wellington Asthma Research Group, Wellington School of Medicine and Health Sciences, University of Otago, Wellington, New Zealand
| | - Christine Barthow
- Wellington Asthma Research Group, Wellington School of Medicine and Health Sciences, University of Otago, Wellington, New Zealand
| | - Edwin A Mitchell
- Department of Paediatrics, Child and Youth Health, The University of Auckland, Auckland, New Zealand
| | - Janice Kang
- Wellington Asthma Research Group, Wellington School of Medicine and Health Sciences, University of Otago, Wellington, New Zealand
| | - Noleen van Zyl
- Department of Paediatrics, Child and Youth Health, The University of Auckland, Auckland, New Zealand
| | - Gordon Purdie
- Dean's Department, Wellington School of Medicine and Health Sciences, University of Otago, Wellington, New Zealand
| | - Thorsten Stanley
- Department of Paediatrics, Wellington School of Medicine and Health Sciences, University of Otago, Wellington, New Zealand
| | | | - Rinki Murphy
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Julian Crane
- Wellington Asthma Research Group, Wellington School of Medicine and Health Sciences, University of Otago, Wellington, New Zealand
| |
Collapse
|
42
|
Slykerman RF, Kang J, Van Zyl N, Barthow C, Wickens K, Stanley T, Coomarasamy C, Purdie G, Murphy R, Crane J, Mitchell EA. Effect of early probiotic supplementation on childhood cognition, behaviour and mood a randomised, placebo-controlled trial. Acta Paediatr 2018; 107:2172-2178. [PMID: 30246890 DOI: 10.1111/apa.14590] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 08/01/2018] [Accepted: 09/20/2018] [Indexed: 12/12/2022]
Abstract
AIM To determine whether probiotic supplementation in early life improves neurocognitive outcomes assessed at 11 years of age. METHODS A total of 474 children who were born March 2004-Aug 2005 participated in a two-centre randomised placebo-controlled trial of infants at risk of developing allergic disease. Pregnant women were randomised to take Lactobacillus rhamnosus strain HN001, Bifidobacterium animalis subsp. lactis strain HN019 or placebo daily from 35 weeks gestation until six months if breastfeeding, and their infants the same treatment from birth to two years. Intelligence, executive function, attention, depression and anxiety were assessed when the children were 11 years of age. RESULTS A total of 342 (72.2%) children were assessed (HN001 n = 109, HN019 n = 118 and placebo n = 115). Overall, there were no significant differences in the neurocognitive outcomes between the treatment groups. CONCLUSION HN001 and HN019 given in early life were not associated with neurocognitive outcomes at 11 years of age in this study. However, we cannot exclude that other probiotics may have a beneficial effect. Further clinical trials are indicated.
Collapse
Affiliation(s)
- R F Slykerman
- Department of Psychological Medicine; University of Auckland; Auckland New Zealand
| | - J Kang
- Department of Medicine; University of Otago; Wellington New Zealand
| | - N Van Zyl
- Department of Paediatrics: Child and Youth Health; University of Auckland; Auckland New Zealand
| | - C Barthow
- Department of Medicine; University of Otago; Wellington New Zealand
| | - K Wickens
- Department of Medicine; University of Otago; Wellington New Zealand
| | - T Stanley
- Department of Paediatrics; University of Otago; Wellington New Zealand
| | - C Coomarasamy
- Department of Paediatrics: Child and Youth Health; University of Auckland; Auckland New Zealand
| | - G Purdie
- Dean's Office; University of Otago; Wellington New Zealand
| | - R Murphy
- Department of Medicine; University of Auckland; Auckland New Zealand
| | - J Crane
- Department of Medicine; University of Otago; Wellington New Zealand
| | - E A Mitchell
- Department of Paediatrics: Child and Youth Health; University of Auckland; Auckland New Zealand
| |
Collapse
|
43
|
Sharma G, Im SH. Probiotics as a Potential Immunomodulating Pharmabiotics in Allergic Diseases: Current Status and Future Prospects. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2018; 10:575-590. [PMID: 30306743 PMCID: PMC6182196 DOI: 10.4168/aair.2018.10.6.575] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/14/2018] [Accepted: 05/21/2018] [Indexed: 01/22/2023]
Abstract
The prevalence of allergic disorders has dramatically increased over the past decade, particularly in developed countries. Apart from gastrointestinal disorders, neoplasia, genital and dermatological diseases etc., dysregulation of gut microbiota (dysbiosis) has also been found to be associated with increased risk of allergies. Probiotics are increasingly being employed to correct dysbiosis and, in turn, to modulate allergic diseases. However, several factors like strain variations and effector metabolites or component of them in a bacterial species can affect the efficacy of those as probiotics. On the other hand, host variations like geographical locations, food habits etc. could also affect the expected results from probiotic usage. Thus, there is a glaring deficiency in our approach to establish probiotics as an irrefutable treatment avenue for suitable disorders. In this review, we explicate on the reported probiotics and their effects on certain allergic diseases like atopic dermatitis, food allergy and asthma to establish their utility. We propose possible measures like elucidation of effector molecules and functional mechanisms of probiotics towards establishing probiotics for therapeutic use. Certain probiotics studies have led to very alarming outcomes which could have been precluded, had effective guidelines been in place. Thus, we also propose ways to secure the safety of probiotics. Overall, our efforts tend to propose necessary discovery and quality assurance guidelines for developing probiotics as potential immunomodulatory 'Pharmabiotics.'
Collapse
Affiliation(s)
- Garima Sharma
- Academy of Immunology and Microbiology (AIM), Institute for Basic Science (IBS), Pohang, Korea
- Division of Integrative Biosciences and Biotechnology, Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Korea
| | - Sin Hyeog Im
- Academy of Immunology and Microbiology (AIM), Institute for Basic Science (IBS), Pohang, Korea
- Division of Integrative Biosciences and Biotechnology, Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Korea.
| |
Collapse
|
44
|
Balato A, Cacciapuoti S, Di Caprio R, Marasca C, Masarà A, Raimondo A, Fabbrocini G. Human Microbiome: Composition and Role in Inflammatory Skin Diseases. Arch Immunol Ther Exp (Warsz) 2018; 67:1-18. [PMID: 30302512 DOI: 10.1007/s00005-018-0528-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 09/03/2018] [Indexed: 12/21/2022]
Abstract
This review focuses on recent evidences about human microbiome composition and functions, exploring the potential implication of its impairment in some diffuse and invalidating inflammatory skin diseases, such as atopic dermatitis, psoriasis, hidradenitis suppurativa and acne. We analysed current scientific literature, focusing on the current evidences about gut and skin microbiome composition and the complex dialogue between microbes and the host. Finally, we examined the consequences of this dialogue for health and skin diseases. This review highlights how human microbes interact with different anatomic niches modifying the state of immune activation, skin barrier status, microbe-host and microbe-microbe interactions. It also shows as most of the factors affecting gut and skin microorganisms' activity have demonstrated to be effective also in modulating chronic inflammatory skin diseases. More and more evidences demonstrate that human microbiome plays a key role in human health and diseases. It is to be expected that these new insights will translate into diagnostic, therapeutic and preventive measures in the context of personalized/precision medicine.
Collapse
Affiliation(s)
- Anna Balato
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, NA, Italy
| | - Sara Cacciapuoti
- Section of Dermatology and Venereology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, NA, Italy.
| | - Roberta Di Caprio
- Section of Dermatology and Venereology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, NA, Italy
| | - Claudio Marasca
- Section of Dermatology and Venereology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, NA, Italy
| | - Anna Masarà
- Section of Dermatology and Venereology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, NA, Italy
| | - Annunziata Raimondo
- Section of Dermatology and Venereology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, NA, Italy
| | - Gabriella Fabbrocini
- Section of Dermatology and Venereology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, NA, Italy
| |
Collapse
|
45
|
Fuglsang E, Krych L, Lundsager MT, Nielsen DS, Frøkiaer H. Postnatal Administration of Lactobacillus rhamnosus HN001 Ameliorates Perinatal Broad-Spectrum Antibiotic-Induced Reduction in Myelopoiesis and T Cell Activation in Mouse Pups. Mol Nutr Food Res 2018; 62:e1800510. [PMID: 30211987 DOI: 10.1002/mnfr.201800510] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 09/03/2018] [Indexed: 12/31/2022]
Abstract
SCOPE This study addresses whether administration of Lactobacillus rhamnosus HN001 could mitigate the effects of a compromised gut microbiota on the composition of mature leukocytes and granulocyte-macrophage progenitor cells (GMPs) in newborn mice. METHODS AND RESULTS Pregnant dams receive oral broad-spectrum antibiotics, which dramatically decrease the gut microbial composition analyzed by 16S rRNA sequencing. Perinatal antibiotic treatment decreases the proportions of bone marrow (BM) GMPs (postnatal day (PND2): 0.5% vs 0.8%, PND4: 0.2% to 0.6%) and mature granulocytes (33% vs 24% at PND2), and spleen granulocytes (7% vs 17% at PND2) and B cells (PND2:18% vs 28%, PND4:11% vs 22%). At PND35, T helper (Th) cells (20% vs 14%) and cytotoxic T (Tc) cells (10% vs 8%) decrease in the spleen. Oral administration of L. rhamnosus HN001 to neonatal pups (PND1-7) restores the antibiotic-induced changes of GMPs and granulocytes in BM and spleen, and further increases splenic granulocytes in control pups. At PND35, splenic proportions of B and Th but not Tc cells are restored. CONCLUSION Postnatal administration of a single bacterial strain efficiently restores granulopoiesis and most T cell activation in neonatal mice that suffer from a perinatal antibiotic-induced compromised gut microbiota at birth.
Collapse
Affiliation(s)
- Eva Fuglsang
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Lukasz Krych
- Department of Food Science, University of Copenhagen, Frederiksberg, Denmark
| | - Mia Thorup Lundsager
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | | | - Hanne Frøkiaer
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
46
|
|
47
|
Hojsak I, Fabiano V, Pop TL, Goulet O, Zuccotti GV, Çokuğraş FC, Pettoello‐Mantovani M, Kolaček S. Guidance on the use of probiotics in clinical practice in children with selected clinical conditions and in specific vulnerable groups. Acta Paediatr 2018; 107:927-937. [PMID: 29446865 PMCID: PMC5969308 DOI: 10.1111/apa.14270] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 01/07/2018] [Accepted: 02/05/2018] [Indexed: 02/05/2023]
Abstract
AIM The use of probiotics has been covered by many guidelines, position papers and evidence-based recommendations, but few have referred to specific patient groups or clinical indications. This review summarises recommendations and scientifically credited guidelines on the use of probiotics for children with selected clinical conditions and provides practice points. METHODS An expert panel was convened by the European Paediatric Association in June 2017 to define the relevant clinical questions for using probiotics in paediatric health care and review and summarise the guidelines, recommendations, position papers and high-quality evidence. RESULTS The panel found that specific probiotic strains were effective in preventing antibiotic-associated and nosocomial diarrhoea, treating acute gastroenteritis and treating infantile colic in breastfed infants. However, special caution is indicated for premature infants, immunocompromised and critically ill patients and those with central venous catheters, cardiac valvular disease and short-gut syndrome. This review discusses the safety of using probiotics in selected groups of paediatric patients and the quality of the available products providing practice points based on proved findings. CONCLUSION Efficacy of probiotics is strain specific. Their benefits are currently scientifically proven for their use in selected clinical conditions in children and not recommended for certain patient groups.
Collapse
Affiliation(s)
- Iva Hojsak
- Children's Hospital ZagrebUniversity of Zagreb School of MedicineZagrebCroatia
- School of MedicineUniversity J.J StrossmayerOsijekCroatia
| | - Valentina Fabiano
- Paediatric DepartmentVittore Buzzi Children's HospitalUniversità degli Studi di MilanoMilanItaly
| | - Tudor Lucian Pop
- 2nd Paediatric ClinicUniversity of Medicine and Pharmacy Iuliu HatieganuCluj‐NapocaRomania
| | - Olivier Goulet
- Department of Paediatric Gastroenterology, Hepatology and NutritionIntestinal Failure Rehabilitation CentreNational Reference Centre for Rare Digestive DiseasesAPHP Necker‐Enfants Malades HospitalParis‐Descartes UniversityParisFrance
| | - Gian Vincenzo Zuccotti
- Paediatric DepartmentVittore Buzzi Children's HospitalUniversità degli Studi di MilanoMilanItaly
| | - Fugen Cullu Çokuğraş
- Paediatric Gastroenterology, Hepatology and NutritionCerrahpaşa Medical FacultyIstanbul UniversityIstanbulTurkey
| | - Massimo Pettoello‐Mantovani
- Department of PaediatricsScientific Institute ‘Casa Sollievo della Sofferenza’University of FoggiaFoggiaItaly
- European Paediatric Association/Union of National European Paediatric Societies and Associations (EPA/UNEPSA)BerlinGermany
| | - Sanja Kolaček
- Children's Hospital ZagrebUniversity of Zagreb School of MedicineZagrebCroatia
| |
Collapse
|
48
|
Wickens K, Barthow C, Mitchell EA, Stanley TV, Purdie G, Rowden J, Kang J, Hood F, van den Elsen L, Forbes-Blom E, Franklin I, Barnes P, Fitzharris P, Maude RM, Stone P, Abels P, Murphy R, Crane J. Maternal supplementation alone with Lactobacillus rhamnosus HN001 during pregnancy and breastfeeding does not reduce infant eczema. Pediatr Allergy Immunol 2018; 29:296-302. [PMID: 29415330 DOI: 10.1111/pai.12874] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/30/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND In a randomized placebo-controlled trial, we previously found that the probiotic Lactobacillus rhamnosus HN001 (HN001) taken by mothers from 35 weeks of gestation until 6 months post-partum if breastfeeding and their child from birth to age 2 years halved the risk of eczema during the first 2 years of life. We aimed to test whether maternal supplementation alone is sufficient to reduce eczema and compare this to our previous study when both the mother and their child were supplemented. METHODS In this 2-centre, parallel double-blind, randomized placebo-controlled trial, the same probiotic as in our previous study (HN001, 6 × 109 colony-forming units) was taken daily by mothers from 14-16 weeks of gestation till 6 months post-partum if breastfeeding, but was not given directly to the child. Women were recruited from the same study population as the first study, where they or their partner had a history of treated asthma, eczema or hay fever. RESULTS Women were randomized to HN001 (N = 212) or placebo (N = 211). Maternal-only HN001 supplementation did not significantly reduce the prevalence of eczema, SCORAD ≥ 10, wheeze or atopic sensitization in the infant by 12 months. This contrasts with the mother and child intervention study, where HN001 was associated with reductions in eczema (hazard ratio (HR): 0.39, 95% CI 0.19-0.79, P = .009) and SCORAD (HR = 0.61, 95% 0.37-1.02). However, differences in the HN001 effect between studies were not significant. HN001 could not be detected in breastmilk from supplemented mothers, and breastmilk TGF-β/IgA profiles were unchanged. CONCLUSION Maternal probiotic supplementation without infant supplementation may not be effective for preventing infant eczema.
Collapse
Affiliation(s)
| | | | | | | | | | - Judy Rowden
- University of Auckland, Auckland, New Zealand
| | - Janice Kang
- University of Otago, Wellington, New Zealand
| | - Fiona Hood
- University of Otago, Wellington, New Zealand
| | | | | | | | | | | | - Robyn M Maude
- Victoria University of Wellington, Wellington, New Zealand
| | - Peter Stone
- University of Auckland, Auckland, New Zealand
| | - Peter Abels
- University of Otago, Wellington, New Zealand
| | | | | |
Collapse
|
49
|
Crane J, Barthow C, Mitchell EA, Stanley TV, Purdie G, Rowden J, Kang J, Hood F, Barnes P, Fitzharris P, Maude R, Stone P, Murphy R, Wickens K. Is yoghurt an acceptable alternative to raw milk for reducing eczema and allergy in infancy? Clin Exp Allergy 2018; 48:604-606. [PMID: 29442385 DOI: 10.1111/cea.13121] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- J Crane
- University of Otago, Wellington, New Zealand
| | - C Barthow
- University of Otago, Wellington, New Zealand
| | | | - T V Stanley
- University of Otago, Wellington, New Zealand
| | - G Purdie
- University of Otago, Wellington, New Zealand
| | - J Rowden
- University of Auckland, Auckland, New Zealand
| | - J Kang
- University of Otago, Wellington, New Zealand
| | - F Hood
- University of Otago, Wellington, New Zealand
| | - P Barnes
- University of Otago, Wellington, New Zealand
| | | | - R Maude
- Victoria University, Wellington, New Zealand
| | - P Stone
- University of Auckland, Auckland, New Zealand
| | - R Murphy
- University of Auckland, Auckland, New Zealand
| | - K Wickens
- University of Otago, Wellington, New Zealand
| |
Collapse
|
50
|
Forsberg A, West CE, Prescott SL, Jenmalm MC. Pre- and probiotics for allergy prevention: time to revisit recommendations? Clin Exp Allergy 2017; 46:1506-1521. [PMID: 27770467 DOI: 10.1111/cea.12838] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Reduced intensity and diversity of microbial exposure is considered a major factor driving abnormal postnatal immune maturation and increasing allergy prevalence, particularly in more affluent regions. Quantitatively, the largest important source of early immune-microbial interaction, the gut microbiota, is of particular interest in this context, with variations in composition and diversity in the first months of life associated with subsequent allergy development. Attempting to restore the health consequences of the 'dysbiotic drift' in modern society, interventions modulating gut microbiota for allergy prevention have been evaluated in several randomized placebo-controlled trials. In this review, we provide an overview of these trials and discuss recommendations from international expert bodies regarding prebiotic, probiotic and synbiotic interventions. Recent guidelines from the World Allergy Organization recommend the use of probiotics for the primary prevention of eczema in pregnant and breastfeeding mothers of infants at high risk for developing allergy and in high-risk infants. It is however stressed that these recommendations are conditional, based on very low-quality evidence and great heterogeneity between studies, which also impedes specific and practical advice to consumers on the most effective regimens. We discuss how the choice of probiotic strains, timing and duration of administration can critically influence the outcome due to different effects on immune modulation and gut microbiota composition. Furthermore, we propose strategies to potentially improve allergy-preventive effects and enable future evidence-based implementation.
Collapse
Affiliation(s)
- A Forsberg
- Division of Neuro and Inflammation Sciences, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - C E West
- International Inflammation (in-FLAME) network of the World Universities Network, Umeå, Sweden.,Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, Sweden
| | - S L Prescott
- International Inflammation (in-FLAME) network of the World Universities Network, Umeå, Sweden.,School of Paediatrics and Child Health, University of Western Australia and Princess Margaret Hospital for Children, Perth, WA, Australia
| | - M C Jenmalm
- Division of Neuro and Inflammation Sciences, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.,International Inflammation (in-FLAME) network of the World Universities Network, Umeå, Sweden
| |
Collapse
|