1
|
Smulders T, Van Der Schee MP, Maitland-Van Der Zee AH, Dikkers FG, Van Drunen CM. Influence of the gut and airway microbiome on asthma development and disease. Pediatr Allergy Immunol 2024; 35:e14095. [PMID: 38451070 DOI: 10.1111/pai.14095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 02/07/2024] [Indexed: 03/08/2024]
Abstract
There are ample data to suggest that early-life dysbiosis of both the gut and/or airway microbiome can predispose a child to develop along a trajectory toward asthma. Although individual studies show clear associations between dysbiosis and asthma development, it is less clear what (collection of) bacterial species is mechanistically responsible for the observed effects. This is partly due to issues related to the asthma diagnosis and the broad spectrum of anatomical sites, sample techniques, and analysis protocols that are used in different studies. Moreover, there is limited attention for potential differences in the genetics of individuals that would affect the outcome of the interaction between the environment and that individual. Despite these challenges, the first bacterial components were identified that are able to affect the transcriptional state of human cells, ergo the immune system. Such molecules could in the future be the basis for intervention studies that are now (necessarily) restricted to a limited number of bacterial species. For this transition, it might be prudent to develop an ex vivo human model of a local mucosal immune system to better and safer explore the impact of such molecules. With this approach, we might move beyond association toward understanding of causality.
Collapse
Affiliation(s)
- Tamar Smulders
- Department of Otorhinolaryngology/Head and Neck Surgery, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Department of Paediatric Pulmonary Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, Netherlands
| | - Marc P Van Der Schee
- Department of Paediatric Pulmonary Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, Netherlands
| | - Anke H Maitland-Van Der Zee
- Department of Paediatric Pulmonary Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, Netherlands
- Department of Pulmonary Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, Netherlands
| | - Frederik G Dikkers
- Department of Otorhinolaryngology/Head and Neck Surgery, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Cornelis M Van Drunen
- Department of Otorhinolaryngology/Head and Neck Surgery, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
2
|
Zaib S, Hayat A, Khan I. Probiotics and their Beneficial Health Effects. Mini Rev Med Chem 2024; 24:110-125. [PMID: 37291788 DOI: 10.2174/1389557523666230608163823] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 05/10/2023] [Accepted: 05/17/2023] [Indexed: 06/10/2023]
Abstract
Probiotics are living microorganisms that are present in cultured milk and fermented food. Fermented foods are a rich source for the isolation of probiotics. They are known as good bacteria. They have various beneficial effects on human health including antihypertensive effects, antihypercholesterolemic effects, prevention of bowel disease, and improving the immune system. Microorganisms including bacteria, yeast, and mold are used as probiotics but the major microorganisms that are used as probiotics are bacteria from the genus Lactobacillus, Lactococcus, Streptococcus, and Bifidobacterium. Probiotics are beneficial in the prevention of harmful effects. Recently, the use of probiotics for the treatment of various oral and skin diseases has also gained significant attention. Clinical studies indicate that the usage of probiotics can alter gut microbiota composition and provoke immune modulation in a host. Due to their various health benefits, probiotics are attaining more interest as a substitute for antibiotics or anti-inflammatory drugs leading to the growth of the probiotic market.
Collapse
Affiliation(s)
- Sumera Zaib
- Department of Basic and Applied Chemistry, Faculty of Sciences and Technology, University of Central Punjab, Lahore, 54590, Pakistan
| | - Aqsa Hayat
- Department of Basic and Applied Chemistry, Faculty of Sciences and Technology, University of Central Punjab, Lahore, 54590, Pakistan
| | - Imtiaz Khan
- Department of Chemistry, Manchester Institute of Biotechnology, The University of Manchester, 131, Princess Street, Manchester M1 7DN, United Kingdom
| |
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
|
Schmid AM, Razim A, Wysmołek M, Kerekes D, Haunstetter M, Kohl P, Brazhnikov G, Geissler N, Thaler M, Krčmářová E, Šindelář M, Weinmayer T, Hrdý J, Schmidt K, Nejsum P, Whitehead B, Palmfeldt J, Schild S, Inić-Kanada A, Wiedermann U, Schabussova I. Extracellular vesicles of the probiotic bacteria E. coli O83 activate innate immunity and prevent allergy in mice. Cell Commun Signal 2023; 21:297. [PMID: 37864211 PMCID: PMC10588034 DOI: 10.1186/s12964-023-01329-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 09/21/2023] [Indexed: 10/22/2023] Open
Abstract
BACKGROUND E. coli O83 (Colinfant Newborn) is a Gram-negative (G-) probiotic bacterium used in the clinic. When administered orally, it reduces allergic sensitisation but not allergic asthma. Intranasal administration offers a non-invasive and convenient delivery method. This route bypasses the gastrointestinal tract and provides direct access to the airways, which are the target of asthma prevention. G- bacteria such as E. coli O83 release outer membrane vesicles (OMVs) to communicate with the environment. Here we investigate whether intranasally administered E. coli O83 OMVs (EcO83-OMVs) can reduce allergic airway inflammation in mice. METHODS EcO83-OMVs were isolated by ultracentrifugation and characterised their number, morphology (shape and size), composition (proteins and lipopolysaccharide; LPS), recognition by innate receptors (using transfected HEK293 cells) and immunomodulatory potential (in naïve splenocytes and bone marrow-derived dendritic cells; BMDCs). Their allergy-preventive effect was investigated in a mouse model of ovalbumin-induced allergic airway inflammation. RESULTS EcO83-OMVs are spherical nanoparticles with a size of about 110 nm. They contain LPS and protein cargo. We identified a total of 1120 proteins, 136 of which were enriched in OMVs compared to parent bacteria. Proteins from the flagellum dominated. OMVs activated the pattern recognition receptors TLR2/4/5 as well as NOD1 and NOD2. EcO83-OMVs induced the production of pro- and anti-inflammatory cytokines in splenocytes and BMDCs. Intranasal administration of EcO83-OMVs inhibited airway hyperresponsiveness, and decreased airway eosinophilia, Th2 cytokine production and mucus secretion. CONCLUSIONS We demonstrate for the first time that intranasally administered OMVs from probiotic G- bacteria have an anti-allergic effect. Our study highlights the advantages of OMVs as a safe platform for the prophylactic treatment of allergy. Video Abstract.
Collapse
Affiliation(s)
- Anna Marlene Schmid
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Agnieszka Razim
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Magdalena Wysmołek
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Daniela Kerekes
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Melissa Haunstetter
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Paul Kohl
- Institute of Molecular Biosciences, Karl-Franzens-University, Graz, Austria
| | - Georgii Brazhnikov
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Nora Geissler
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Michael Thaler
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Eliška Krčmářová
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University, and General University Hospital, Prague, Czech Republic
| | - Martin Šindelář
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Tamara Weinmayer
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Jiří Hrdý
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University, and General University Hospital, Prague, Czech Republic
| | - Katy Schmidt
- Core Facility for Cell Imaging and Ultrastructural Research, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Peter Nejsum
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Bradley Whitehead
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Johan Palmfeldt
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Stefan Schild
- Institute of Molecular Biosciences, Karl-Franzens-University, Graz, Austria
- BioTechMed, Graz, Austria
- Field of Excellence Biohealth - University of Graz, Graz, Austria
| | - Aleksandra Inić-Kanada
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Ursula Wiedermann
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Irma Schabussova
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria.
| |
Collapse
|
5
|
Kummola L, González-Rodríguez MI, Marnila P, Nurminen N, Salomaa T, Hiihtola L, Mäkelä I, Laitinen OH, Hyöty H, Sinkkonen A, Junttila IS. Comparison of the effect of autoclaved and non-autoclaved live soil exposure on the mouse immune system : Effect of soil exposure on immune system. BMC Immunol 2023; 24:29. [PMID: 37689649 PMCID: PMC10492337 DOI: 10.1186/s12865-023-00565-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 08/29/2023] [Indexed: 09/11/2023] Open
Abstract
BACKGROUND . Lack of exposure to the natural microbial diversity of the environment has been linked to dysregulation of the immune system and numerous noncommunicable diseases, such as allergies and autoimmune disorders. Our previous studies suggest that contact with soil material, rich in naturally occurring microbes, could have a beneficial immunoregulatory impact on the immune system in mice and humans. However, differences in the immunomodulatory properties of autoclaved, sterile soil material and non-autoclaved, live soil material have not been compared earlier. RESULTS . In this study, we exposed C57BL/6 mice to autoclaved and live soil powders that had the same rich microbiota before autoclaving. We studied the effect of the soil powders on the mouse immune system by analyzing different immune cell populations, gene expression in the gut, mesenteric lymph nodes and lung, and serum cytokines. Both autoclaved and live soil exposure were associated with changes in the immune system. The exposure to autoclaved soil resulted in higher levels of Rorγt, Inos and Foxp3 expression in the colon. The exposure to live soil was associated with elevated IFN-γ concentration in the serum. In the mesenteric lymph node, exposure to live soil reduced Gata3 and Foxp3 expression, increased the percentage of CD8 + T cells and the expression of activation marker CD80 in XCR1+SIRPα- migratory conventional dendritic cell 1 subset. CONCLUSIONS . Our results indicate that exposure to the live and autoclaved soil powders is not toxic for mice. Exposure to live soil powder slightly skews the immune system towards type 1 direction which might be beneficial for inhibiting type 2-related inflammation. Further studies are warranted to quantify the impact of this exposure in experimental type 2 inflammation.
Collapse
Affiliation(s)
- Laura Kummola
- Faculty of Medicine and Health Technology, Tampere University, Tampere, 33014, Finland
| | | | - Pertti Marnila
- Natural Resources Institute Finland (Luke), Jokioinen, Finland
| | - Noora Nurminen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, 33014, Finland
| | - Tanja Salomaa
- Faculty of Medicine and Health Technology, Tampere University, Tampere, 33014, Finland
- Fimlab Laboratories, Arvo-Building, Rm F326, Arvo Ylpön katu 34, Tampere, 33520, Finland
| | - Lotta Hiihtola
- Faculty of Medicine and Health Technology, Tampere University, Tampere, 33014, Finland
- Fimlab Laboratories, Arvo-Building, Rm F326, Arvo Ylpön katu 34, Tampere, 33520, Finland
| | - Iida Mäkelä
- Natural Resources Institute Finland (Luke), Jokioinen, Finland
| | - Olli H Laitinen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, 33014, Finland
| | - Heikki Hyöty
- Faculty of Medicine and Health Technology, Tampere University, Tampere, 33014, Finland
| | - Aki Sinkkonen
- Natural Resources Institute Finland (Luke), Jokioinen, Finland
| | - Ilkka S Junttila
- Faculty of Medicine and Health Technology, Tampere University, Tampere, 33014, Finland.
- Fimlab Laboratories, Arvo-Building, Rm F326, Arvo Ylpön katu 34, Tampere, 33520, Finland.
- Northern Finland Laboratory Centre (NordLab), Oulu, 90220, Finland.
- Research Unit of Biomedicine, University of Oulu, Oulu, 90570, Finland.
| |
Collapse
|
6
|
Zheng H, Dai H, Yan X, Xiang Q. Study on Intestinal Flora and Asthma: Knowledge Graph Analysis Based on CiteSpace (2001-2021). J Asthma Allergy 2023; 16:355-364. [PMID: 37041761 PMCID: PMC10083019 DOI: 10.2147/jaa.s402883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 03/12/2023] [Indexed: 04/13/2023] Open
Abstract
Asthma is a common chronic inflammatory disease of the airway. Intestinal flora, a significant risk factor for asthma, has become a widespread concern in the pathogenesis of asthma. To review the literature related to intestinal flora in asthma, summarize research direction, and report trends, this study used CiteSpace to perform bibliometric statistics and analysis on the research papers of intestinal flora and asthma collected in the Web of science core collection from 2001 to 2021. Eventually, a total of 613 articles were included. The results demonstrated that research on gut flora and asthma continued to heat up, with article numbers increasing, especially in the last decade. Moreover, analysis of the keywords showed that the research topics of intestinal flora and asthma range from confirming the link between intestinal flora and asthma to investigating mechanisms and then to asthma treatment. According to the summary of research hotspots, we expand on three emerging issues that require attention in the intestinal flora and asthma research, including (regulatory T)Treg cells, probiotics, and chain fatty acid. Evidence illustrated that Treg cells play a crucial role in the pathogenesis of asthma caused by dysbiosis of the gut flora. Furthermore, in contrast to probiotic supplements, which do not reduce the risk of developing asthma, short-chain fatty acids supplements do. Overall, the research direction in the field of intestinal flora and asthma has recently evolved from macro to micro with depth broadened. As a robust scientific evaluation, our study provided a comprehensive overview of the area, particularly for research focus, which could more precisely direct scholars on future research and clinical diagnosis, therapy, and individualized prevention.
Collapse
Affiliation(s)
- Hang Zheng
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Huan Dai
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Xiumei Yan
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Qiangwei Xiang
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
- Correspondence: Qiangwei Xiang, Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China, Email
| |
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
|
Fiocchi A, Cabana MD, Mennini M. Current Use of Probiotics and Prebiotics in Allergy. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2022; 10:2219-2242. [PMID: 35792336 DOI: 10.1016/j.jaip.2022.06.038] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 06/27/2022] [Accepted: 06/27/2022] [Indexed: 01/06/2023]
Abstract
The microbiome plays an important role in the pathogenesis of allergic diseases. This review updates the reader on studies aimed at influencing allergic diseases through modulation of the gut microflora. A nonsystematic review of the literature was performed, focusing on relevant trials evaluating the effect of probiotics/prebiotics/symbiotics in the prevention and treatment of allergic disease. For each allergic disease, we were able to find not only a substantial number of clinical trials but also systematic reviews. Specific guidelines, based on systematic reviews and meta-analyses, are available for the prevention of allergic disease and for the treatment of food allergy. In each of the areas examined-allergic rhinitis, allergic asthma, atopic dermatitis, food allergy, and gastrointestinal allergies-there are substantial uncertainties in the efficacy of gut microflora modulation in prevention and treatment. At present, practicing clinicians can avail themselves of intestinal flora modulators as an adjunct in the prevention of atopic dermatitis but not of other forms of allergic diseases. Their effects on the treatment of allergic diseases remain controversial.
Collapse
Affiliation(s)
- Alessandro Fiocchi
- Translational Research in Pediatric Specialities Area, Allergy Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
| | - Michael D Cabana
- Department of Pediatrics, Albert Einstein College of Medicine and the Children's Hospital at Montefiore, Bronx, NY
| | - Maurizio Mennini
- Translational Research in Pediatric Specialities Area, Allergy Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
10
|
Chen N, Liu F, Gao Q, Wang R, Zhang L, Li Y. A Meta-Analysis of Probiotics for the Treatment of Allergic Airway Diseases in Children and Adolescents. Am J Rhinol Allergy 2022; 36:480-490. [PMID: 35238209 DOI: 10.1177/19458924221080159] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Allergic airway disease is a chronic airway allergic inflammatory disease including allergic rhinitis (AR) and allergic asthma which is common in children and adolescents. Recently the probiotics has been becoming a supplementary or alternative therapy to allergic diseases, however the effect of them has not been clearly established. OBJECTIVE The purpose of the present meta-analysis was to evaluate the effectiveness of probiotics on allergic airway disease including AR and allergic asthma in children and adolescents. METHODS We performed a comprehensive search on PubMed, Cochrane Library, EMBASE for relevant publications from 1 Jan 2000 to 1 July 2021. Physical examinations, Pediatric Rhinoconjunctivitis Quality of Life Questionnaires (PRQLQs), Total Nasal Symptom Score (TNSS), Nasal or Eye Symptom Score (NSS or ESS), serum allergen-specific IgE, and eosinophil were used as evaluating indicators for AR and allergic asthma in children and adolescents. The meta-analysis was performed using Review Manager (RevMan, Version 5.3). RESULTS 15 randomized controlled trials (RCTs) with a total of 1388 participants were included for the meta-analysis. Among them, 729 patients treated with probiotics served as the probiotics group, and 659 patients with placebo as control group. Significantly greater reduction in PRQLQs from baseline to endpoint (SMD = -2.57, 95% CI [ - 4.66, -0.48] P < 0.01), NSS (SMD = -1.43, 95% CI [ - 1.63, - 1.23], P < 0.01) and ESS (total MD = -1.67, 95% CI [ - 1.79, - 1.55], P < 0.01) were observed in probiotics group compared to control group. Probiotics have no significant effect to serum IgE and eosinophils (P > 0.01). CONCLUSION The results of this meta-analysis indicated that probiotics treatment may reduce PRQLQs, NSS, ESS in patients with allergic airway disease. More research involving the mechanism of probiotics are needed to clarify the role of probiotics in AR and allergic asthma in children and adolescents.
Collapse
Affiliation(s)
- Na Chen
- Department of Otolaryngology, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Fang Liu
- Department of Otolaryngology, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Qiang Gao
- Department of Gastroenterology and Hepatology, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Rui Wang
- Department of Otolaryngology, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Lei Zhang
- Department of Otolaryngology, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Youwei Li
- Department of Radiology, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
11
|
Probiotics in Children with Asthma. CHILDREN 2022; 9:children9070978. [PMID: 35883962 PMCID: PMC9316460 DOI: 10.3390/children9070978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [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
|
12
|
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: 33] [Impact Index Per Article: 11.0] [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
|
13
|
Satia I, Cusack R, Stevens C, Schlatman A, Wattie J, Mian F, Killian KJ, O'Byrne PM, Bienenstock J, Forsythe P, Gauvreau GM. Limosilactobacillus reuteri DSM-17938 for preventing cough in adults with mild allergic asthma: A double-blind randomized placebo-controlled cross-over study. Clin Exp Allergy 2021; 51:1133-1143. [PMID: 34192396 DOI: 10.1111/cea.13976] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/27/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Cough is a common troublesome symptom in asthma which is neuronally mediated. Limosilactobacillus reuteri DSM-17938 (L. reuteri DSM-17938) is a probiotic shown to be effective in pre-clinical models at suppressing neuronal responses to capsaicin, a transient receptor potential vanilloid agonist (TRPV1). OBJECTIVE Investigate the effects of DSM-17938 versus matched placebo on capsaicin-evoked coughs in mild allergic asthmatics. METHODS We performed a 4-visit, randomized, double-blind, placebo-controlled, two-way cross-over study comparing full dose cough responses with inhaled capsaicin in mild allergic asthmatics after 1 month of treatment with DSM-17938 compared with matched placebo. Randomization and allocation to trial group were carried out by a central computer system. Histamine skin prick testing, airway hyper-responsiveness and inflammatory cells in induced sputum were measured at every visit. Blood was collected to extract PBMCs and stimulated with CD3/CD28 to ascertain the effects of DSM-17938 /placebo on T-cell cytokine responses. RESULTS Seventeen subjects were recruited and 15 completed the study (8 females, mean age 27.3 years). There was no difference in the change in maximum capsaicin-evoked coughs (Emax) after treatment with L. reuteri DSM-17938 compared with placebo [mean difference 2.07 coughs (95% CI -2.77 to 6.91, p = .38) or relative changes in geometric mean ratios for the dose evoking at least half the Emax (ED50) [1.05 (95% CI 0.31-3.58, p = .94)], concentration evoking 2 coughs (C2) [0.63 (0.26-1.53), p = .28] and 5 coughs (C5) [0.79 (0.25-2.50), p = .67]. There was no effect on histamine skin prick wheal size, intensity of itch sensation, methacholine PC20, airway inflammation or T-cell responses after stimulation with CD3/CD28. There were no serious adverse events. One subject developed a mild upper respiratory tract infection and another mild transient nausea whilst on DSM-17938. CONCLUSION In this small study in adults with mild allergic asthma, we found no evidence that L. reuteri DSM-17938 has any systemic effects on airway nerves, smooth muscle, sputum inflammatory cells, skin responses or T-cell responses after oral consumption. TRIAL REGISTRATION Clinicaltrials.gov Identifier: NCT03603522.
Collapse
Affiliation(s)
- Imran Satia
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada.,Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, ON, Canada
| | - Ruth Cusack
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Catie Stevens
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Abbey Schlatman
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Jennifer Wattie
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Firoz Mian
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Kieran J Killian
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Paul M O'Byrne
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada.,Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, ON, Canada
| | - John Bienenstock
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Paul Forsythe
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Gail M Gauvreau
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
14
|
Jakubczyk D, Górska S. Impact of Probiotic Bacteria on Respiratory Allergy Disorders. Front Microbiol 2021; 12:688137. [PMID: 34234762 PMCID: PMC8256161 DOI: 10.3389/fmicb.2021.688137] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/28/2021] [Indexed: 12/22/2022] Open
Abstract
Respiratory allergy is a common disease with an increased prevalence worldwide. The effective remedy is still unknown, and a new therapeutic approach is highly desirable. The review elaborates the influence of probiotic bacteria on respiratory allergy prevention and treatment with particular emphasis on the impact of the current methods of their administration – oral and intranasal. The background of the respiratory allergy is complex thus, we focused on the usefulness of probiotics in the alleviation of different allergy factors, in particular involved in pathomechanism, local hypersensitive evidence and the importance of epithelial barrier. In this review, we have shown that (1) probiotic strains may vary in modulatory potential in respiratory allergy, (2) probiotic bacteria are beneficial in oral and intranasal administration, (3) recombinant probiotic bacteria can modulate the course of respiratory allergy.
Collapse
Affiliation(s)
- Dominika Jakubczyk
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Sabina Górska
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| |
Collapse
|
15
|
Casaro MB, Thomas AM, Mendes E, Fukumori C, Ribeiro WR, Oliveira FA, Crisma AR, Murata GM, Bizzarro B, Sá-Nunes A, Setubal JC, Mayer MPA, Martins FS, Vieira AT, Antiorio ATFB, Tavares-de-Lima W, Camara NOS, Curi R, Dias-Neto E, Ferreira CM. A probiotic has differential effects on allergic airway inflammation in A/J and C57BL/6 mice and is correlated with the gut microbiome. MICROBIOME 2021; 9:134. [PMID: 34112246 PMCID: PMC8194189 DOI: 10.1186/s40168-021-01081-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 04/20/2021] [Indexed: 06/12/2023]
Abstract
The phenotypes of allergic airway diseases are influenced by the interplay between host genetics and the gut microbiota, which may be modulated by probiotics. We investigated the probiotic effects on allergic inflammation in A/J and C57BL/6 mice. C57BL/6 mice had increased gut microbiota diversity compared to A/J mice at baseline. Acetate producer probiotics differentially modulated and altered the genus abundance of specific bacteria, such as Akkermansia and Allistipes, in mouse strains. We induced airway inflammation followed by probiotic treatment and found that only A/J mice exhibited decreased inflammation, and the beneficial effects of probiotics in A/J mice were partially due to acetate production. To understand the relevance of microbial composition colonization in the development of allergic diseases, we implanted female C57BL/6 mice with A/J embryos to naturally modulate the microbial composition of A/J mice, which increased gut microbiota diversity and reduced eosinophilic inflammation in A/J. These data demonstrate the central importance of microbiota to allergic phenotype severity. Video Abstract.
Collapse
Affiliation(s)
- Mateus B Casaro
- Department of Pharmaceutics Sciences, Institute of Environmental, Chemistry and Pharmaceutical Sciences, Universidade Federal de São Paulo, R. São Nicolau, 210, Diadema, SP, 09913-03, Brazil
| | - Andrew M Thomas
- Department CIBIO, University of Trento, Trento, Italy
- Medical Genomics Laboratory, CIPE/A.C. Camargo Cancer Center, São Paulo, Brazil
- Department of Biochemistry, Institute of Chemistry, Universidade de São Paulo, São Paulo, Brazil
| | - Eduardo Mendes
- Department of Pharmaceutics Sciences, Institute of Environmental, Chemistry and Pharmaceutical Sciences, Universidade Federal de São Paulo, R. São Nicolau, 210, Diadema, SP, 09913-03, Brazil
| | - Claudio Fukumori
- Department of Pharmaceutics Sciences, Institute of Environmental, Chemistry and Pharmaceutical Sciences, Universidade Federal de São Paulo, R. São Nicolau, 210, Diadema, SP, 09913-03, Brazil
| | - Willian R Ribeiro
- Department of Pharmaceutics Sciences, Institute of Environmental, Chemistry and Pharmaceutical Sciences, Universidade Federal de São Paulo, R. São Nicolau, 210, Diadema, SP, 09913-03, Brazil
| | - Fernando A Oliveira
- Center for Mathematics, Computing and Cognition (CMCC), Federal University of ABC - UFABC, São Bernardo do Campo, SP, Brazil
| | - Amanda R Crisma
- Department of Clinical Analyses, Universidade Federal do Paraná, Curitiba, Brazil
| | - Gilson M Murata
- Department of Medical Clinic, Faculty of Medicine, University of São Paulo, São Paulo, 01246-903, Brazil
| | - Bruna Bizzarro
- Department of Immunology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo, Brazil
| | - Anderson Sá-Nunes
- Department of Immunology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo, Brazil
| | - Joao C Setubal
- Department of Biochemistry, Institute of Chemistry, Universidade de São Paulo, São Paulo, Brazil
| | - Marcia P A Mayer
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Flaviano S Martins
- Department of Microbiology, Institute of Biological Sciences, Federal Universidade de Minas Gerais, Belo Horizonte, Brazil
| | - Angélica T Vieira
- Department of Biochemistry and Immunology, Biological Science Institute, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Ana T F B Antiorio
- Department of Pathology, School of Veterinary Medicine and Animal Science, Universidade de São Paulo, São Paulo, Brazil
| | - Wothan Tavares-de-Lima
- Department of Pharmacology, Institute of Biomedical Sciences I, Universidade de São Paulo, São Paulo, Brazil
| | - Niels O S Camara
- Department of Immunology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo, Brazil
| | - Rui Curi
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo, Brazil
| | - Emmanuel Dias-Neto
- Medical Genomics Laboratory, CIPE/A.C. Camargo Cancer Center, São Paulo, Brazil
- Laboratory of Neurosciences (LIM-27), Institute of Psychiatry, Medical School, Universidade de São Paulo, São Paulo, Brazil
| | - Caroline M Ferreira
- Department of Pharmaceutics Sciences, Institute of Environmental, Chemistry and Pharmaceutical Sciences, Universidade Federal de São Paulo, R. São Nicolau, 210, Diadema, SP, 09913-03, Brazil.
| |
Collapse
|
16
|
Setiawan EA, Rianda D, Kadim M, Meilianawati, Susanto F, Kok FJ, Shankar AH, Agustina R. Tenth year reenrollment randomized trial investigating the effects of childhood probiotics and calcium supplementation on height and weight at adolescence. Sci Rep 2021; 11:11860. [PMID: 34088920 PMCID: PMC8178317 DOI: 10.1038/s41598-021-88819-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 04/06/2021] [Indexed: 02/05/2023] Open
Abstract
Microbiota and its modification with specific probiotics in early life could provide long term health benefits. Probiotics and calcium strengthen intestinal integrity and may support linear growth. This study investigated the long-term effects of childhood probiotics and calcium supplementation on growth in adolescence. We re-enrolled 238 adolescents aged 11-18 years from 494 children 10-years after 6-months of supplementation with either low-lactose milk fortified with low levels of calcium (LC, ∼50 mg/day, n = 53/124), with regular levels of calcium (RC, ∼440 mg/day, n = 70/126), or with regular calcium + 5 x 108 CFU/day Lactobacillus reuteri DSM 17938 (Reuteri, n = 55/124), or regular calcium + 5 x 108 CFU/day L. casei CRL 431 (Casei, n = 60/120). Changes in height-for-age z-score (HAZ) and body mass index-for-age z-score (BMIZ) were determined from the end of intervention to re-enrollment. General linear models were used to assess the effects on HAZ and BMIZ of group, gender, living area, maternal education, family income, physical activity, diet quality, nutritional status, and gut integrity as determined by urinary lactulose/mannitol ratio (L:M). Adolescent mean age was 15.3 years, mean HAZ was - 1.11, mean BMIZ was - 0.2 and median L:M (n = 155) was 0.23. Changes in HAZ and BMIZ were not significantly different between Casei, Reuteri, LC compared to RC. However, a significant decrease in BMIZ was observed among female adolescents in the Casei compared to RC group (- 0.5 SD, 95% CI - 0.8 to - 0.003, p = 0.048). Childhood probiotic and calcium supplementation may therefore selectively affect female adolescents.Clinical trial registration: This follow-up study has been registered at www.clinicaltrials.gov , Registry name: Rina Agustina, Registration number: NCT04046289, First Registration Date 06/08/19. web link: https://www.clinicaltrials.gov/ct2/show/NCT04046289 .
Collapse
Affiliation(s)
- Evania Astella Setiawan
- Department of Nutrition, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo General Hospital, Jl. Salemba Raya No.6, Jakarta, 10430, Indonesia
| | - Davrina Rianda
- Department of Nutrition, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo General Hospital, Jl. Salemba Raya No.6, Jakarta, 10430, Indonesia
- Human Nutrition Research Center, Indonesian Medical Education and Research Institute (HNRC-IMERI), Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Muzal Kadim
- Department of Pediatric, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Meilianawati
- Department of Nutrition, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo General Hospital, Jl. Salemba Raya No.6, Jakarta, 10430, Indonesia
| | - Fenny Susanto
- Department of Nutrition, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo General Hospital, Jl. Salemba Raya No.6, Jakarta, 10430, Indonesia
| | - Frans J Kok
- Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Anuraj H Shankar
- Human Nutrition Research Center, Indonesian Medical Education and Research Institute (HNRC-IMERI), Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Eijkman-Oxford Clinical Research Unit, Eijkman Institute for Molecular Biology, Jakarta, Indonesia
| | - Rina Agustina
- Department of Nutrition, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo General Hospital, Jl. Salemba Raya No.6, Jakarta, 10430, Indonesia.
- Human Nutrition Research Center, Indonesian Medical Education and Research Institute (HNRC-IMERI), Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.
| |
Collapse
|
17
|
Sun S, Chang G, Zhang L. The prevention effect of probiotics against eczema in children: an update systematic review and meta-analysis. J DERMATOL TREAT 2021; 33:1844-1854. [PMID: 34006167 DOI: 10.1080/09546634.2021.1925077] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Accumulated evidences support the fetus's intestinal flora unbalance is associated with the development of allergic diseases. Probiotic supplements in pregnancy and childhood might prevent atopic diseases. The aim of this systematic review and meta-analysis was to evaluate the effect of probiotic supplementation during pregnancy and early infancy in preventing eczema, atopic eczema, and other allergic diseases. We also explored whether different probiotic strains or intervention objects affected the antiallergic effect of probiotics and the prevention atopy effect of the long-term period. Fixed-effect models were used, and random-effects models where significant heterogeneity was present. Results were expressed as odds ratios (ORs) with a 95% confidence interval (CI). Twenty-one studies were included in the meta-analysis. The probiotics group had a significantly lower risk of eczema and atopic eczema compared to controls, especially those treated with probiotic combinations. Mothers' probiotics intake significantly contributed to reducing the risk of eczema as well as atopic eczema. What's more, probiotics seemed effective on eczema prevention ≤2 years of age, but against atopic eczema after 1 of age year. No significant difference in terms of prevention of asthma, rhinitis, wheeze, allergic diseases and sensation. In brief, a probiotic supplement is expected to become a novel potential strategy for infant eczema and atopic eczema.
Collapse
Affiliation(s)
- Shuya Sun
- Graduate school, Tianjin Medical University, Tianjin, China
| | - Guizhen Chang
- Department of Dermatology, Tianjin TEDA Hospital, Tianjin, China
| | - Litao Zhang
- Department of Dermatology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
| |
Collapse
|
18
|
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 2021; 8:860-875. [PMID: 32147139 DOI: 10.1016/j.jaip.2019.12.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [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
|
19
|
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: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/23/2021] [Accepted: 04/24/2021] [Indexed: 02/07/2023] Open
Abstract
Asthma is a major global health issue. Over 300 million people worldwide suffer from this chronic inflammatory airway disease. Typical clinical symptoms of asthma are characterized by a recurrent wheezy cough, chest tightness, and shortness of breath. The main goals of asthma management are to alleviate asthma symptoms, reduce the risk of asthma exacerbations, and minimize long-term medicinal adverse effects. However, currently available type 2 T helper cells (Th2)-directed treatments are often ineffective due to the heterogeneity of the asthma subgroups, which manifests clinically with variable and poor treatment responses. Personalized precision therapy of asthma according to individualized clinical characteristics (phenotype) and laboratory biomarkers (endotype) is the future prospect. This mini review discusses the molecular mechanisms underlying asthma pathogenesis, including the hot sought-after topic of microbiota, add-on therapies and the potential application of probiotics in the management of asthma.
Collapse
Affiliation(s)
- Chiao-Juno Chiu
- Graduate Institute of Clinical Medicine, School of Medicine, National Taiwan University, Taipei 100, Taiwan;
| | - Miao-Tzu Huang
- Graduate Institute of Clinical Medicine, School of Medicine, National Taiwan University, Taipei 100, Taiwan;
- Department of Medical Research, National Taiwan University Hospital, No. 7 Chung-Shan South Road, Taipei 100, Taiwan
- Department of Pediatrics, National Taiwan University Hospital, No. 7 Chung-Shan South Road, Taipei 100, Taiwan
| |
Collapse
|
20
|
Raza GS, Herzig KH, Leppäluoto J. Invited review: Milk fat globule membrane-A possible panacea for neurodevelopment, infections, cardiometabolic diseases, and frailty. J Dairy Sci 2021; 104:7345-7363. [PMID: 33896625 DOI: 10.3168/jds.2020-19649] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 02/15/2021] [Indexed: 12/23/2022]
Abstract
Milk is an evolutionary benefit for humans. For infants, it offers optimal nutrients for normal growth, neural development, and protection from harmful microbes. Humans are the only mammals who drink milk throughout their life. Lipids in colostrum originate mostly from milk fat globule membrane (MFGM) droplets extruded from the mammary gland. The MFGM gained much interest as a potential nutraceutical, due to their high phospholipid (PL), ganglioside (GD), and protein contents. In this review, we focused on health effects of MFGM ingredients and dairy food across the life span, especially on neurodevelopment, cardiometabolic health, and frailty in older adults. The MFGM supplements to infants and children reduced gastrointestinal and respiratory tract infections and improved neurodevelopment due to the higher content of protein, PL, and GD in MFGM. The MFGM formulas containing PL and GD improved brain myelination and fastened nerve conduction speed, resulting in improved behavioral developments. Administration of MFGM-rich ingredients improved insulin sensitivity and decreased inflammatory markers, LDL-cholesterol, and triglycerides by lowering intestinal absorption of cholesterol and increasing its fecal excretion. The MFGM supplements, together with exercise, improved ambulatory activities, leg muscle mass, and muscle fiber velocity in older adults. There are great variations in the composition of lipids and proteins in MFGM products, which make comparisons of the different studies impossible. In addition, investigations of the individual MFGM components are required to evaluate their specific effects and molecular mechanisms. Although we are currently only beginning to understand the possible health effects of MFGM products, the current MFGM supplementation trials as presented in this review have shown significant clinical health benefits across the human life span, which are worth further investigation.
Collapse
Affiliation(s)
- Ghulam Shere Raza
- Research Unit of Biomedicine, Medical Research Center, Faculty of Medicine, University of Oulu, 90014 Oulu, Finland
| | - Karl-Heinz Herzig
- Research Unit of Biomedicine, Medical Research Center, Faculty of Medicine, University of Oulu, 90014 Oulu, Finland; Oulu University Hospital, 90220 Oulu, Finland; Pediatric Institute, Poznan University of Medical Sciences, 60-572 Poznan, Poland
| | - Juhani Leppäluoto
- Research Unit of Biomedicine, Medical Research Center, Faculty of Medicine, University of Oulu, 90014 Oulu, Finland.
| |
Collapse
|
21
|
Stavropoulou E, Bezirtzoglou E. Probiotics in Medicine: A Long Debate. Front Immunol 2020; 11:2192. [PMID: 33072084 PMCID: PMC7544950 DOI: 10.3389/fimmu.2020.02192] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 08/11/2020] [Indexed: 12/12/2022] Open
Abstract
During the last years probiotics gained the attention of clinicians for their use in the prevention and treatment of multiple diseases. Probiotics main mechanisms of action include enhanced mucosal barrier function, direct antagonism with pathogens, inhibition of bacterial adherence and invasion capacity in the intestinal epithelium, boosting of the immune system and regulation of the central nervous system. It is accepted that there is a mutual communication between the gut microbiota and the liver, the so-called “microbiota-gut-liver axis” as well as a reciprocal communication between the intestinal microbiota and the central nervous system through the “microbiota-gut-brain axis.” Moreover, recently the “gut-lung axis” in bacterial and viral infections is considerably discussed for bacterial and viral infections, as the intestinal microbiota amplifies the alveolar macrophage activity having a protective role in the host defense against pneumonia. The importance of the normal human intestinal microbiota is recognized in the preservation of health. Disease states such as, infections, autoimmune conditions, allergy and other may occur when the intestinal balance is disturbed. Probiotics seem to be a promising approach to prevent and even reduce the symptoms of such clinical states as an adjuvant therapy by preserving the balance of the normal intestinal microbiota and improving the immune system. The present review states globally all different disorders in which probiotics can be given. To date, Stronger data in favor of their clinical use are provided in the prevention of gastrointestinal disorders, antibiotic-associated diarrhea, allergy and respiratory infections. We hereby discuss the role of probiotics in the reduction of the respiratory infection symptoms and we focus on the possibility to use them as an adjuvant to the therapeutic approach of the pandemic COVID-19. Nevertheless, it is accepted by the scientific community that more clinical studies should be undertaken in large samples of diseased populations so that the assessment of their therapeutic potential provide us with strong evidence for their efficacy and safety in clinical use.
Collapse
Affiliation(s)
- Elisavet Stavropoulou
- CHUV (Centre Hospitalier Universitaire Vaudois), Lausanne, Switzerland.,Department of Infectious Diseases, Central Institute, Valais Hospital, Sion, Switzerland
| | - Eugenia Bezirtzoglou
- Laboratory of Hygiene and Environmental Protection, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| |
Collapse
|
22
|
Navarro-Tapia E, Sebastiani G, Sailer S, Toledano LA, Serra-Delgado M, García-Algar Ó, Andreu-Fernández V. Probiotic Supplementation During the Perinatal and Infant Period: Effects on Gut Dysbiosis and Disease. Nutrients 2020; 12:E2243. [PMID: 32727119 PMCID: PMC7468726 DOI: 10.3390/nu12082243] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/15/2020] [Accepted: 07/22/2020] [Indexed: 02/07/2023] Open
Abstract
The perinatal period is crucial to the establishment of lifelong gut microbiota. The abundance and composition of microbiota can be altered by several factors such as preterm delivery, formula feeding, infections, antibiotic treatment, and lifestyle during pregnancy. Gut dysbiosis affects the development of innate and adaptive immune responses and resistance to pathogens, promoting atopic diseases, food sensitization, and infections such as necrotizing enterocolitis (NEC). Recent studies have indicated that the gut microbiota imbalance can be restored after a single or multi-strain probiotic supplementation, especially mixtures of Lactobacillus and Bifidobacterium strains. Following the systematic search methodology, the current review addresses the importance of probiotics as a preventive or therapeutic tool for dysbiosis produced during the perinatal and infant period. We also discuss the safety of the use of probiotics in pregnant women, preterm neonates, or infants for the treatment of atopic diseases and infections.
Collapse
Affiliation(s)
- Elisabet Navarro-Tapia
- Grup de Recerca Infancia i Entorn (GRIE), Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Valencian International University (VIU), 46002 Valencia, Spain
| | - Giorgia Sebastiani
- Department of Neonatology, Hospital Clínic-Maternitat, ICGON, BCNatal, 08028 Barcelona, Spain
| | - Sebastian Sailer
- Department of Neonatology, Hospital Clínic-Maternitat, ICGON, BCNatal, 08028 Barcelona, Spain
| | - Laura Almeida Toledano
- Institut de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
- BCNatal, Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), University of Barcelona, 08950 Barcelona, Spain
| | - Mariona Serra-Delgado
- Institut de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
- BCNatal, Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), University of Barcelona, 08950 Barcelona, Spain
| | - Óscar García-Algar
- Grup de Recerca Infancia i Entorn (GRIE), Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Department of Neonatology, Hospital Clínic-Maternitat, ICGON, BCNatal, 08028 Barcelona, Spain
| | - Vicente Andreu-Fernández
- Grup de Recerca Infancia i Entorn (GRIE), Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Valencian International University (VIU), 46002 Valencia, Spain
- Department of Neonatology, Hospital Clínic-Maternitat, ICGON, BCNatal, 08028 Barcelona, Spain
| |
Collapse
|
23
|
Forsberg A, Huoman J, Söderholm S, Bhai Mehta R, Nilsson L, Abrahamsson TR, Ernerudh J, Gustafsson M, Jenmalm MC. Pre- and postnatal Lactobacillus reuteri treatment alters DNA methylation of infant T helper cells. Pediatr Allergy Immunol 2020; 31:544-553. [PMID: 32150651 DOI: 10.1111/pai.13240] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 12/20/2019] [Accepted: 01/27/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Perinatal childhood exposures, including probiotic supplementation, may affect epigenetic modifications and impact on immune maturation and allergy development. The aim of this study was to assess the effects of pre- and postnatal Lactobacillus reuteri supplementation on DNA methylation in relation to immune maturation and allergy development. METHODS DNA methylation patterns were investigated for allergy-related T helper subsets using a locus-specific method and at a genome-wide scale using the Illumina 450K array. From a randomised, double-blind, placebo-controlled allergy prevention trial with pre- and postnatal probiotic supplementation, CD4+ T helper cells were obtained at birth (from cord blood), and 12 and 24 months of age (total (placebo/probiotics); locus-specific method: CB = 32 (17/15), 12 months = 24 (9/15), 24 months = 35 (15/20); Illumina: CB = 19 (10/9), 12 months = 10 (6/4), 24 months = 19(11/8)). RESULTS Comparing probiotics to placebo, the greatest genome-wide differential DNA methylation was observed at birth, where the majority of sites were hypomethylated, indicating transcriptional accessibility in the probiotic group. Bioinformatic analyses, including network analyses, revealed a module containing 91 genes, enriched for immune-related pathways such as chemotaxis, PI3K-Akt, MAPK and TGF-β signalling. A majority of the module genes were associated with atopic manifestations (OR = 1.43, P = 2.4 × 10-6 ), and a classifier built on this model could predict allergy development (AUC = 0.78, P = 3.0 × 10e-3 ). Pathways such as IFN-γ signalling and T-cell activation were more hypermethylated at birth compared with later in life in both intervention groups over time, in line with DNA methylation patterns in the IFNG locus obtained by the locus-specific methodology. CONCLUSION Maternal L. reuteri supplementation during pregnancy alters DNA methylation patterns in CD4+ T cells towards enhanced immune activation at birth, which may affect immune maturation and allergy development.
Collapse
Affiliation(s)
- Anna Forsberg
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Johanna Huoman
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Simon Söderholm
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Wallenberg Centre for Molecular Medicine, Linköping University, Linköping, Sweden
| | - Ratnesh Bhai Mehta
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Lennart Nilsson
- Allergy Center, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Thomas R Abrahamsson
- Crown Princess Victoria's Child and Youth Hospital, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Jan Ernerudh
- Department of Clinical Immunology and Transfusion Medicine, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Mika Gustafsson
- Department of Physics, Chemistry and Biology, Bioinformatics, Linköping University, Linköping, Sweden
| | - Maria C Jenmalm
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
24
|
Oral and nasal probiotic administration for the prevention and alleviation of allergic diseases, asthma and chronic obstructive pulmonary disease. Nutr Res Rev 2020; 34:1-16. [PMID: 32281536 DOI: 10.1017/s0954422420000116] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Interaction between a healthy microbiome and the immune system leads to body homeostasis, as dysbiosis in microbiome content and loss of diversity may result in disease development. Due to the ability of probiotics to help and modify microbiome constitution, probiotics are now widely used for the prevention and treatment of different gastrointestinal, inflammatory, and, more recently, respiratory diseases. In this regard, chronic respiratory diseases including chronic obstructive pulmonary disease (COPD), asthma and allergic rhinitis are among the most common and complicated respiratory diseases with no specific treatment until now. Accordingly, many studies have evaluated the therapeutic efficacy of probiotic administration (mostly via the oral route and much lesser nasal route) on chronic respiratory diseases. We tried to summarise and evaluate these studies to give a perspective of probiotic therapy via both the oral and nasal routes for respiratory infections (in general) and chronic respiratory diseases (specifically). We finally concluded that probiotics might be useful for allergic diseases. For asthmatic patients, probiotics can modulate serum cytokines and IgE and decrease eosinophilia, but with no significant reduction in clinical symptoms. For COPD, only limited studies were found with uncertain clinical efficacy. For intranasal administration, although some studies propose more efficiency than the oral route, more clinical evaluations are warranted.
Collapse
|
25
|
Dzidic M, Mira A, Artacho A, Abrahamsson TR, Jenmalm MC, Collado MC. Allergy development is associated with consumption of breastmilk with a reduced microbial richness in the first month of life. Pediatr Allergy Immunol 2020; 31:250-257. [PMID: 31736150 DOI: 10.1111/pai.13176] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 11/08/2019] [Accepted: 11/12/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND Early colonization with a diverse microbiota seems to play a crucial role for appropriate immune maturation during childhood. Breastmilk microbiota is one important source of microbes for the infant, transferred together with maternal IgA antibodies. We previously observed that allergy development during childhood was associated with aberrant IgA responses to the gut microbiota already at 1 month of age, when the IgA antibodies are predominantly maternally derived in breastfed infants. OBJECTIVE To determine the microbial composition and IgA-coated bacteria in breastmilk in relation to allergy development in children participating in an intervention trial with pre- and post-natal Lactobacillus reuteri supplementation. METHODS A combination of flow cytometric cell sorting and 16S rRNA gene sequencing was used to characterize the bacterial recognition patterns by IgA in breastmilk samples collected one month post-partum from 40 mothers whose children did or did not develop allergic and asthmatic symptoms during the first 7 years of age. RESULTS The milk fed to children developing allergic manifestations had significantly lower bacterial richness, when compared to the milk given to children that remained healthy. Probiotic treatment influenced the breastmilk microbiota composition. However, the proportions of IgA-coated bacteria, the total bacterial load and the patterns of IgA-coating were similar in breastmilk between mothers of healthy children and those developing allergies. CONCLUSION Consumption of breastmilk with a reduced microbial richness in the first month of life may play an important role in allergy development during childhood.
Collapse
Affiliation(s)
- Majda Dzidic
- Department of Biotechnology, Institute of Agrochemistry and Food Technology (IATA-CSIC), Unit of Lactic Acid Bacteria and Probiotics, Valencia, Spain.,Department of Health and Genomics, Center for Advanced Research in Public Health, FISABIO, Valencia, Spain.,Department of Clinical and Experimental Medicine, Division of Autoimmunity and Immune Regulation, Linköping University, Linköping, Sweden
| | - Alex Mira
- Department of Health and Genomics, Center for Advanced Research in Public Health, FISABIO, Valencia, Spain.,CIBER-ESP, Madrid, Spain
| | - Alejandro Artacho
- Department of Health and Genomics, Center for Advanced Research in Public Health, FISABIO, Valencia, Spain
| | - Thomas R Abrahamsson
- Department of Clinical and Experimental Medicine and Department of Pediatrics, Linköping University, Linköping, Sweden
| | - Maria C Jenmalm
- Department of Clinical and Experimental Medicine, Division of Autoimmunity and Immune Regulation, Linköping University, Linköping, Sweden
| | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology (IATA-CSIC), Unit of Lactic Acid Bacteria and Probiotics, Valencia, Spain
| |
Collapse
|
26
|
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: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 11/20/2019] [Indexed: 11/19/2022] Open
|
27
|
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: 11] [Impact Index Per Article: 2.2] [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
|
28
|
Shukla SD, Shastri MD, Chong WC, Dua K, Budden KF, Mahmood MQ, Hansbro NG, Keely S, Eri R, Patel RP, Peterson GM, Hansbro PM. Microbiome-focused asthma management strategies. Curr Opin Pharmacol 2019; 46:143-149. [PMID: 31357048 DOI: 10.1016/j.coph.2019.06.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 06/24/2019] [Accepted: 06/24/2019] [Indexed: 12/11/2022]
Abstract
Asthma is a common, heterogeneous and serious disease with high prevalence globally. Poorly controlled, steroid-resistant asthma is particularly important as there are no effective therapies and it exerts substantial healthcare and societal burden. The role of microbiomes, particularly in chronic diseases has generated considerable interest in recent times. Existing evidence clearly demonstrates an association between asthma initiation and the microbiome, both respiratory and gastro-intestinal, although its' roles are poorly understood when assessing the asthma progression or heterogeneity (i.e. phenotypes/endotypes) across different geographical locations. Moreover, modulating microbiomes could be preventive and/or therapeutic in patients with asthma warrants urgent attention. Here, we review recent advances in assessing the role of microbiomes in asthma and present the challenges associated with the potential therapeutic utility of modifying microbiomes in management.
Collapse
Affiliation(s)
- Shakti D Shukla
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute & University of Newcastle, Callaghan, NSW, Australia
| | - Madhur D Shastri
- School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Australia
| | - Wai Chin Chong
- Department of Molecular and Translational Science, Monash University, Clayton, Australia; Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Australia
| | - Kamal Dua
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute & University of Newcastle, Callaghan, NSW, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, Australia
| | - Kurtis F Budden
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute & University of Newcastle, Callaghan, NSW, Australia
| | - Malik Quasir Mahmood
- Medicine, College of Health and Medicine, University of Tasmania, Hobart, Australia
| | - Nicole G Hansbro
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute & University of Newcastle, Callaghan, NSW, Australia; Centre for inflammation, Centenary Institute, Sydney, and School of Life Sciences, University of Technology, Ultimo, NSW, Australia
| | - Simon Keely
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute & University of Newcastle, Callaghan, NSW, Australia
| | - Rajaraman Eri
- School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Australia
| | - Rahul P Patel
- Pharmacy, College of Health and Medicine, University of Tasmania, Hobart, Australia
| | - Gregory M Peterson
- Pharmacy, College of Health and Medicine, University of Tasmania, Hobart, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute & University of Newcastle, Callaghan, NSW, Australia; Centre for inflammation, Centenary Institute, Sydney, and School of Life Sciences, University of Technology, Ultimo, NSW, Australia.
| |
Collapse
|
29
|
Li L, Han Z, Niu X, Zhang G, Jia Y, Zhang S, He C. Probiotic Supplementation for Prevention of Atopic Dermatitis in Infants and Children: A Systematic Review and Meta-analysis. Am J Clin Dermatol 2019; 20:367-377. [PMID: 30465329 DOI: 10.1007/s40257-018-0404-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Probiotic supplementation in early life may be effective in preventing atopic dermatitis (AD); however, results regarding efficacy have been controversial. OBJECTIVE The aim of our study was to investigate the effect of probiotic supplementation on the risk of AD. METHODS We systematically searched PubMed, EBSCO, Embase and Web of Science databases up to 8 March 2018 for potentially relevant studies regarding probiotic supplementation and AD. Included infants and children were those with probiotic exposure in utero and/or after birth who were not previously diagnosed with AD. We calculated the odds ratios (ORs) and 95% confidence intervals (CIs) and used the Jadad and Newcastle-Ottawa scales to assess methodologic quality. RESULTS A total of 28 studies met the inclusion criteria. Compared with controls, probiotic treatment was associated with a reduced risk of AD (OR 0.69; 95% CI 0.58-0.82, P < 0.0001). The use of probiotics during both the prenatal and the postnatal period significantly reduced the incidence of AD (OR 0.67; 95% CI 0.54-0.82); however, analysis of studies of probiotics given prenatally only or postnatally only did not reach statistical significance. CONCLUSIONS Our meta-analysis showed that probiotic supplementation during both the prenatal and the postnatal period reduced the incidence of AD in infants and children. Our findings suggest that starting probiotic treatment during gestation and continuing through the first 6 months of the infant's life may be of benefit in the prevention of AD.
Collapse
Affiliation(s)
- Lin Li
- Department of Gastroenterology, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, Anhui, People's Republic of China
| | - Zhen Han
- Department of Gastroenterology, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, Anhui, People's Republic of China
| | - Xiaoping Niu
- Department of Gastroenterology, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, Anhui, People's Republic of China
| | - Guozheng Zhang
- Department of Gastroenterology, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, Anhui, People's Republic of China
| | - Yuliang Jia
- Department of Gastroenterology, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, Anhui, People's Republic of China
| | - Shunguo Zhang
- Department of Gastroenterology, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, Anhui, People's Republic of China
| | - Chiyi He
- Department of Gastroenterology, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, Anhui, People's Republic of China.
| |
Collapse
|
30
|
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: 53] [Impact Index Per Article: 10.6] [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
|
31
|
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: 30] [Impact Index Per Article: 6.0] [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
|
32
|
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: 3.7] [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
|
33
|
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: 49] [Impact Index Per Article: 8.2] [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
|
34
|
Dzidic M, Abrahamsson TR, Artacho A, Collado MC, Mira A, Jenmalm MC. Oral microbiota maturation during the first 7 years of life in relation to allergy development. Allergy 2018; 73:2000-2011. [PMID: 29602225 DOI: 10.1111/all.13449] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2018] [Indexed: 12/26/2022]
Abstract
BACKGROUND Allergic diseases have become a major public health problem in affluent societies. Microbial colonization early in life seems to be critical for instructing regulation on immune system maturation and allergy development in children. Even though the oral cavity is the first site of encounter between a majority of foreign antigens and the immune system, the influence of oral bacteria on allergy development has not yet been reported. OBJECTIVE We sought to determine the bacterial composition in longitudinally collected saliva samples during childhood in relation to allergy development. METHODS Illumina sequencing of the 16S rDNA gene was used to characterize the oral bacterial composition in saliva samples collected at 3, 6, 12, 24 months, and 7 years of age from children developing allergic symptoms and sensitization (n = 47) and children staying healthy (n = 33) up to 7 years of age. RESULTS Children developing allergic disease, particularly asthma, had lower diversity of salivary bacteria together with highly divergent bacterial composition at 7 years of age, showing a clearly altered oral microbiota in these individuals, likely as a consequence of an impaired immune system during infancy. Moreover, the relative amounts of several bacterial species, including increased abundance of Gemella haemolysans in children developing allergies and Lactobacillus gasseri and L. crispatus in healthy children, were distinctive during early infancy, likely influencing early immune maturation. CONCLUSION Early changes in oral microbial composition seem to influence immune maturation and allergy development. Future experiments should test the probiotic potential of L. gasseri and L. crispatus isolates.
Collapse
Affiliation(s)
- M. Dzidic
- Department of Biotechnology Unit of Lactic Acid Bacteria and Probiotics Institute of Agrochemistry and Food Technology (IATA‐CSIC) Valencia Spain
- Department of Health and Genomics Center for Advanced Research in Public Health, FISABIO Valencia Spain
- CIBER‐ESP Madrid Spain
- Division of Autoimmunity and Immune Regulation Department of Clinical and Experimental Medicine Linköping University Linköping Sweden
| | - T. R. Abrahamsson
- Department of Clinical and Experimental Medicine Division of Pediatrics Linköping University Linköping Sweden
| | - A. Artacho
- Department of Health and Genomics Center for Advanced Research in Public Health, FISABIO Valencia Spain
- CIBER‐ESP Madrid Spain
| | - M. C. Collado
- Department of Biotechnology Unit of Lactic Acid Bacteria and Probiotics Institute of Agrochemistry and Food Technology (IATA‐CSIC) Valencia Spain
| | - A. Mira
- Department of Health and Genomics Center for Advanced Research in Public Health, FISABIO Valencia Spain
- CIBER‐ESP Madrid Spain
| | - M. C. Jenmalm
- Division of Autoimmunity and Immune Regulation Department of Clinical and Experimental Medicine Linköping University Linköping Sweden
| |
Collapse
|
35
|
Dzidic M, Collado MC, Abrahamsson T, Artacho A, Stensson M, Jenmalm MC, Mira A. Oral microbiome development during childhood: an ecological succession influenced by postnatal factors and associated with tooth decay. THE ISME JOURNAL 2018; 12:2292-2306. [PMID: 29899505 PMCID: PMC6092374 DOI: 10.1038/s41396-018-0204-z] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 05/08/2018] [Accepted: 05/23/2018] [Indexed: 12/24/2022]
Abstract
Information on how the oral microbiome develops during early childhood and how external factors influence this ecological process is scarce. We used high-throughput sequencing to characterize bacterial composition in saliva samples collected at 3, 6, 12, 24 months and 7 years of age in 90 longitudinally followed children, for whom clinical, dietary and health data were collected. Bacterial composition patterns changed through time, starting with "early colonizers", including Streptococcus and Veillonella; other bacterial genera such as Neisseria settled after 1 or 2 years of age. Dental caries development was associated with diverging microbial composition through time. Streptococcus cristatus appeared to be associated with increased risk of developing tooth decay and its role as potential biomarker of the disease should be studied with species-specific probes. Infants born by C-section had initially skewed bacterial content compared with vaginally delivered infants, but this was recovered with age. Shorter breastfeeding habits and antibiotic treatment during the first 2 years of age were associated with a distinct bacterial composition at later age. The findings presented describe oral microbiota development as an ecological succession where altered colonization pattern during the first year of life may have long-term consequences for child´s oral and systemic health.
Collapse
Affiliation(s)
- Majda Dzidic
- Department of Health and Genomics, Center for Advanced Research in Public Health, CSISP-FISABIO, Valencia, Spain
- Institute of Agrochemistry and Food Technology (IATA-CSIC), Department of Biotechnology, Unit of Lactic Acid Bacteria and Probiotics, Valencia, Spain
- Department of Clinical and Experimental Medicine, Division of Autoimmunity and Immune Regulation, Linköping University, Linköping, Sweden
| | - Maria C Collado
- Institute of Agrochemistry and Food Technology (IATA-CSIC), Department of Biotechnology, Unit of Lactic Acid Bacteria and Probiotics, Valencia, Spain
| | - Thomas Abrahamsson
- Department of Clinical and Experimental Medicine, Division of Pediatrics, Linköping University, Linköping, Sweden
| | - Alejandro Artacho
- Department of Health and Genomics, Center for Advanced Research in Public Health, CSISP-FISABIO, Valencia, Spain
| | - Malin Stensson
- Centre for Oral Health, School of Health and Welfare, Jönköping University, Jönköping, Sweden
| | - Maria C Jenmalm
- Department of Clinical and Experimental Medicine, Division of Autoimmunity and Immune Regulation, Linköping University, Linköping, Sweden
| | - Alex Mira
- Department of Health and Genomics, Center for Advanced Research in Public Health, CSISP-FISABIO, Valencia, Spain.
| |
Collapse
|
36
|
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: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [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 Zagreb; University of Zagreb School of Medicine; Zagreb Croatia
- School of Medicine; University J.J Strossmayer; Osijek Croatia
| | - Valentina Fabiano
- Paediatric Department; Vittore Buzzi Children's Hospital; Università degli Studi di Milano; Milan Italy
| | - Tudor Lucian Pop
- 2nd Paediatric Clinic; University of Medicine and Pharmacy Iuliu Hatieganu; Cluj-Napoca Romania
| | - Olivier Goulet
- Department of Paediatric Gastroenterology, Hepatology and Nutrition; Intestinal Failure Rehabilitation Centre; National Reference Centre for Rare Digestive Diseases; APHP Necker-Enfants Malades Hospital; Paris-Descartes University; Paris France
| | - Gian Vincenzo Zuccotti
- Paediatric Department; Vittore Buzzi Children's Hospital; Università degli Studi di Milano; Milan Italy
| | - Fugen Cullu Çokuğraş
- Paediatric Gastroenterology, Hepatology and Nutrition; Cerrahpaşa Medical Faculty; Istanbul University; Istanbul Turkey
| | - Massimo Pettoello-Mantovani
- Department of Paediatrics; Scientific Institute ‘Casa Sollievo della Sofferenza’; University of Foggia; Foggia Italy
- European Paediatric Association/Union of National European Paediatric Societies and Associations (EPA/UNEPSA); Berlin Germany
| | - Sanja Kolaček
- Children's Hospital Zagreb; University of Zagreb School of Medicine; Zagreb Croatia
| |
Collapse
|
37
|
Grönroos M, Parajuli A, Laitinen OH, Roslund MI, Vari HK, Hyöty H, Puhakka R, Sinkkonen A. Short-term direct contact with soil and plant materials leads to an immediate increase in diversity of skin microbiota. Microbiologyopen 2018; 8:e00645. [PMID: 29808965 PMCID: PMC6436432 DOI: 10.1002/mbo3.645] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/23/2018] [Accepted: 03/27/2018] [Indexed: 12/14/2022] Open
Abstract
Immune‐mediated diseases have increased during the last decades in urban environments. The hygiene hypothesis suggests that increased hygiene level and reduced contacts with natural biodiversity are related to the increase in immune‐mediated diseases. We tested whether short‐time contact with microbiologically diverse nature‐based materials immediately change bacterial diversity on human skin. We tested direct skin contact, as two volunteers rubbed their hands with sixteen soil and plant based materials, and an exposure via fabric packets filled with moss material. Skin swabs were taken before and after both exposures. Next‐generation sequencing showed that exposures increased, at least temporarily, the total diversity of skin microbiota and the diversity of Acidobacteria, Actinobacteria, Bacteroidetes, Proteobacteria and Alpha‐, Beta‐ and Gammaproteobacteria suggesting that contact with nature‐based materials modify skin microbiome and increase skin microbial diversity. Until now, approaches to cure or prevent immune system disorders using microbe‐based treatments have been limited to use of a few microbial species. We propose that nature‐based materials with high natural diversity, such as the materials tested here, might be more effective in modifying human skin microbiome, and eventually, in reducing immune system disorders. Future studies should investigate how long‐term changes in skin microbiota are achieved and if the exposure induces beneficial changes in the immune system markers.
Collapse
Affiliation(s)
- Mira Grönroos
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Lahti, Finland
| | - Anirudra Parajuli
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Lahti, Finland
| | - Olli H Laitinen
- Department of Virology, School of Medicine, University of Tampere, Tampere, Finland
| | - Marja I Roslund
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Lahti, Finland
| | - Heli K Vari
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Lahti, Finland
| | - Heikki Hyöty
- Department of Virology, School of Medicine, University of Tampere, Tampere, Finland.,Fimlab Laboratories, Pirkanmaa Hospital District, Tampere, Finland
| | - Riikka Puhakka
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Lahti, Finland
| | - Aki Sinkkonen
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Lahti, Finland
| |
Collapse
|
38
|
Mu Q, Tavella VJ, Luo XM. Role of Lactobacillus reuteri in Human Health and Diseases. Front Microbiol 2018; 9:757. [PMID: 29725324 PMCID: PMC5917019 DOI: 10.3389/fmicb.2018.00757] [Citation(s) in RCA: 403] [Impact Index Per Article: 67.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 04/04/2018] [Indexed: 12/13/2022] Open
Abstract
Lactobacillus reuteri (L. reuteri) is a well-studied probiotic bacterium that can colonize a large number of mammals. In humans, L. reuteri is found in different body sites, including the gastrointestinal tract, urinary tract, skin, and breast milk. The abundance of L. reuteri varies among different individuals. Several beneficial effects of L. reuteri have been noted. First, L. reuteri can produce antimicrobial molecules, such as organic acids, ethanol, and reuterin. Due to its antimicrobial activity, L. reuteri is able to inhibit the colonization of pathogenic microbes and remodel the commensal microbiota composition in the host. Second, L. reuteri can benefit the host immune system. For instance, some L. reuteri strains can reduce the production of pro-inflammatory cytokines while promoting regulatory T cell development and function. Third, bearing the ability to strengthen the intestinal barrier, the colonization of L. reuteri may decrease the microbial translocation from the gut lumen to the tissues. Microbial translocation across the intestinal epithelium has been hypothesized as an initiator of inflammation. Therefore, inflammatory diseases, including those located in the gut as well as in remote tissues, may be ameliorated by increasing the colonization of L. reuteri. Notably, the decrease in the abundance of L. reuteri in humans in the past decades is correlated with an increase in the incidences of inflammatory diseases over the same period of time. Direct supplementation or prebiotic modulation of L. reuteri may be an attractive preventive and/or therapeutic avenue against inflammatory diseases.
Collapse
Affiliation(s)
| | | | - Xin M. Luo
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
39
|
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: 6.6] [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
|
40
|
West CE, Dzidic M, Prescott SL, Jenmalm MC. Bugging allergy; role of pre-, pro- and synbiotics in allergy prevention. Allergol Int 2017; 66:529-538. [PMID: 28865967 DOI: 10.1016/j.alit.2017.08.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 08/02/2017] [Accepted: 08/02/2017] [Indexed: 02/07/2023] Open
Abstract
Large-scale biodiversity loss and complex changes in social behaviors are altering human microbial ecology. This is increasingly implicated in the global rise in inflammatory diseases, most notably the "allergy epidemic" in very early life. Colonization of human ecological niches, particularly the gastrointestinal tract, is critical for normal local and systemic immune development and regulation. Disturbances in composition, diversity and timing of microbial colonization have been associated with increased allergy risk, indicating the importance of strategies to restore a dysbiotic gut microbiota in the primary prevention of allergic diseases, including the administration of probiotics, prebiotics and synbiotics. Here, we summarize and discuss findings of randomized clinical trials that have examined the effects of these microbiome-related strategies on short and long-term allergy preventative effects - including new guidelines from the World Allergy Organization which now recommend probiotics and prebiotics for allergy prevention under certain conditions. The relatively low quality evidence, limited comparative studies and large heterogeneity between studies, have collectively hampered recommendations on specific probiotic strains, specific timing and specific conditions for the most effective preventive management. At the same time the risk of using available products is low. While further research is needed before specific practice guidelines on supplement probiotics and prebiotics, it is equally important that the underlying dietary and lifestyle factors of dysbiosis are addressed at both the individual and societal levels.
Collapse
Affiliation(s)
- Christina E West
- Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, Sweden; inFLAME Global Network (Worldwide Universities Network), West New York, NJ, USA.
| | - Majda Dzidic
- inFLAME Global Network (Worldwide Universities Network), West New York, NJ, USA; Division of Neuro and Inflammation Sciences, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden; Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Department of Biotechnology, Unit of Lactic Acid Bacteria and Probiotics, Valencia, Spain
| | - Susan L Prescott
- inFLAME Global Network (Worldwide Universities Network), West New York, NJ, USA; School of Paediatrics and Child Health, University of Western Australia and Princess Margaret Hospital for Children, Perth, Australia
| | - Maria C Jenmalm
- inFLAME Global Network (Worldwide Universities Network), West New York, NJ, USA; Division of Neuro and Inflammation Sciences, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
41
|
McAleer JP, Kolls JK. Contributions of the intestinal microbiome in lung immunity. Eur J Immunol 2017; 48:39-49. [PMID: 28776643 DOI: 10.1002/eji.201646721] [Citation(s) in RCA: 153] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 06/12/2017] [Accepted: 08/01/2017] [Indexed: 12/20/2022]
Abstract
The intestine is a critical site of immune cell development that not only controls intestinal immunity but extra-intestinal immunity as well. Recent findings have highlighted important roles for gut microbiota in shaping lung inflammation. Here, we discuss interactions between the microbiota and immune system including T cells, protective effects of microbiota on lung infections, the role of diet in shaping the composition of gut microbiota and susceptibility to asthma, epidemiologic evidence implicating antibiotic use and microbiota in asthma and clinical trials investigating probiotics as potential treatments for atopy and asthma. The systemic effects of gut microbiota are partially attributed to their generating metabolites including short chain fatty acids, which can suppress lung inflammation through the activation of G protein-coupled receptors. Thus, studying the interactions between microbiota and immune cells can lead to the identification of therapeutic targets for chronic lower respiratory diseases.
Collapse
Affiliation(s)
- Jeremy P McAleer
- Department of Pharmaceutical Science and Research, Marshall University School of Pharmacy, Huntington, WV, USA
| | - Jay K Kolls
- Tulane School of Medicine, Center for Translational Research in Infection and Inflammation, New Orleans, LA, USA
| |
Collapse
|
42
|
Prevention of allergies in childhood - where are we now? Allergol Select 2017; 1:200-213. [PMID: 30402617 PMCID: PMC6040005 DOI: 10.5414/alx01807e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 07/27/2015] [Indexed: 11/18/2022] Open
Abstract
Allergic diseases represent an increasing health problem for children worldwide. Along with allergic airway diseases, food allergy comes to the fore and herewith closely intertwined the hypothesis that an early allergic sensitization might occur via skin barrier defect(s). The importance of the skin barrier has been documented by several studies meanwhile. Not only genetic studies screen the associations between Filaggrin loss-of-function mutations, atopic dermatitis, allergic sensitization, food allergy and even airway diseases, but also epidemiological studies cast new light on the hypothesis of the atopic march. As another focus in context of the development of an allergic phenotype, the specific microbial exposure with all its diversities has been crystallized as it shapes the immune system in (early) infancy. Studies explored both, the role of human intestinal microbiota as well as the external microbial diversity. Unfortunately suitable markers for atopic predictors are still rare. New studies point out that specific IgE antibodies (e.g., IgE to Phl p 1) in children without allergic symptoms so far, might function as a pre-clinical biomarker, which may help to identify candidates for primary (allergen non-specific) or secondary (allergen-specific) prevention in terms of specific immunoprophylaxis. These manifold research activities document a complex increase in knowledge. Nevertheless new assumptions need to be substantively confirmed in order to finally generate the urgently needed preventive strategies for allergic diseases in childhood.
Collapse
|
43
|
Early exposure to agricultural soil accelerates the maturation of the early-life pig gut microbiota. Anaerobe 2017; 45:31-39. [DOI: 10.1016/j.anaerobe.2017.02.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 02/23/2017] [Accepted: 02/24/2017] [Indexed: 12/27/2022]
|
44
|
Peldan P, Kukkonen AK, Savilahti E, Kuitunen M. Perinatal probiotics decreased eczema up to 10 years of age, but at 5-10 years, allergic rhino-conjunctivitis was increased. Clin Exp Allergy 2017; 47:975-979. [PMID: 28316095 DOI: 10.1111/cea.12924] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- P Peldan
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - A K Kukkonen
- Skin and Allergy Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - E Savilahti
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - M Kuitunen
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
45
|
Lee-Sarwar KA, Bacharier LB, Litonjua AA. Strategies to alter the natural history of childhood asthma. Curr Opin Allergy Clin Immunol 2017; 17:139-145. [PMID: 28079559 PMCID: PMC5664210 DOI: 10.1097/aci.0000000000000340] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW Asthma exhibits significant heterogeneity in occurrence and severity over the lifespan. Our goal is to discuss recent evidence regarding determinants of the natural history of asthma during childhood, and review the rationale behind and status of major efforts to alter its course. RECENT FINDINGS Variations in microbial exposures are associated with risk of allergic disease, and the use of bacterial lysates may be a promising preventive strategy. Exposure to air pollution appears to be particularly damaging in prenatal and early life, and interventions to reduce pollution are feasible and result in clinical benefit. E-cigarette use may have a role in harm reduction for conventional cigarette smokers with asthma, but has undefined short-term and long-term effects that must be clarified. Vitamin D insufficiency over the first several years of life is associated with risk of asthma, and vitamin D supplementation reduces the risk of severe exacerbations. SUMMARY The identification of risk factors for asthma occurrence, persistence and severity will continue to guide efforts to alter the natural history of the disease. We have reviewed several promising strategies that are currently under investigation. Vitamin D supplementation and air pollution reduction have been shown to be effective strategies and warrant increased investigation and implementation.
Collapse
Affiliation(s)
- K A Lee-Sarwar
- aDivision of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, Massachusetts bDivision of Pediatric Allergy, Immunology and Pulmonary Medicine, Department of Pediatrics, Washington University School of Medicine cSt Louis Children's Hospital, St Louis, Missouri dChanning Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | | |
Collapse
|
46
|
Gray LEK, O'Hely M, Ranganathan S, Sly PD, Vuillermin P. The Maternal Diet, Gut Bacteria, and Bacterial Metabolites during Pregnancy Influence Offspring Asthma. Front Immunol 2017; 8:365. [PMID: 28408909 PMCID: PMC5374203 DOI: 10.3389/fimmu.2017.00365] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 03/14/2017] [Indexed: 01/04/2023] Open
Abstract
This review focuses on the current evidence that maternal dietary and gut bacterial exposures during pregnancy influence the developing fetal immune system and subsequent offspring asthma. Part 1 addresses exposure to a farm environment, antibiotics, and prebiotic and probiotic supplementation that together indicate the importance of bacterial experience in immune programming and offspring asthma. Part 2 outlines proposed mechanisms to explain these associations including bacterial exposure of the fetoplacental unit; immunoglobulin-related transplacental transport of gut bacterial components; cytokine signaling producing fetomaternal immune alignment; and immune programming via metabolites produced by gut bacteria. Part 3 focuses on the interplay between diet, gut bacteria, and bacterial metabolites. Maternal diet influences fecal bacterial composition, with dietary microbiota-accessible carbohydrates (MACs) selecting short-chain fatty acid (SCFA)-producing bacteria. Current evidence from mouse models indicates an association between increased maternal dietary MACs, SCFA exposure during pregnancy, and reduced offspring asthma that is, at least in part, mediated by the induction of regulatory T lymphocytes in the fetal lung. Part 4 discusses considerations for future studies investigating maternal diet-by-microbiome determinants of offspring asthma including the challenge of measuring dietary MAC intake; limitations of the existing measures of the gut microbiome composition and metabolic activity; measures of SCFA exposure; and the complexities of childhood respiratory health assessment.
Collapse
Affiliation(s)
- Lawrence E K Gray
- Barwon Infant Study, School of Medicine, Deakin University, Geelong, VIC, Australia.,Child Health Research Unit, Barwon Health, Geelong, VIC, Australia
| | - Martin O'Hely
- Barwon Infant Study, School of Medicine, Deakin University, Geelong, VIC, Australia.,Respiratory Diseases, Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia
| | - Sarath Ranganathan
- Respiratory Diseases, Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Respiratory and Sleep Medicine, Royal Children's Hospital, Parkville, VIC, Australia.,Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Peter David Sly
- Child Health Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Peter Vuillermin
- Barwon Infant Study, School of Medicine, Deakin University, Geelong, VIC, Australia.,Child Health Research Unit, Barwon Health, Geelong, VIC, Australia
| |
Collapse
|
47
|
Ho YH, Huang YT, Lu YC, Lee SY, Tsai MF, Hung SP, Hsu TY. Effects of Gender and Age on Immune Responses of Human Peripheral Blood Mononuclear Cells to Probiotics: A Large Scale Pilot Study. J Nutr Health Aging 2017; 21:521-526. [PMID: 28448082 DOI: 10.1007/s12603-016-0818-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Despite the widely accepted concept that probiotics confer miscellaneous benefits to hosts, the controversies surrounding these health-promoting claims cannot be ignored. These controversies hinder development and innovation in this field. RESULTS To clarify the effects of age and gender on probiotic-induced immune responses, we recruited 1613 Taiwanese individuals and calculated the ratio of IFN-γ to IL-10 production after each individual's PBMCs were stimulated by six probiotic strains (L. paracasei BRAP01, L. acidophilus AD300, B. longum BA100, E. faecium BR0085, L. rhamnosus AD500 and L. reuteri BR101). Our results indicated that gender and age have only minor effects on the immune modulation of probiotics. Additionally, we showed that L. paracasei BRAP01 and L. acidophilus AD300 are the two dominant strains inducing IFN-γ/IL-10 production in Taiwanese individuals and that L. reuteri BR101 was the most effective stimulator of IL-10/IFN-γ. Additionally, a significant inverse relationship between the ability of L. paracasei BRAP01 and L. rhamnosus AD500 to stimulate IFN-γ/IL-10 or IL-10/IFN-γ production was also observed. CONCLUSIONS Our results indicated that age and gender have only minor effects on the immune modulation abilities of probiotics.
Collapse
Affiliation(s)
- Y-H Ho
- Ting-Yuan Hsu, MD, Ph.D. 5F., No.466, Bo'ai 1st Rd., Gushan Dist., Kaohsiung City 80466, Taiwan (R.O.C.), Phone: +886-7-5579268 ext. 510 , E-mail:
| | | | | | | | | | | | | |
Collapse
|
48
|
Mennini M, Dahdah L, Artesani MC, Fiocchi A, Martelli A. Probiotics in Asthma and Allergy Prevention. Front Pediatr 2017; 5:165. [PMID: 28824889 PMCID: PMC5534455 DOI: 10.3389/fped.2017.00165] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Accepted: 07/13/2017] [Indexed: 12/16/2022] Open
Abstract
Interest in probiotic research and its potential benefits in infant foods are relatively recent but significantly increasing. The evolution of the knowledge in the last 20 years demonstrated that alterations in the microbiome may be a consequence of events occurring during infancy or childhood, including prematurity, cesarean section, and nosocomial infections. Several pieces of evidence prove that a "healthy" intestinal microbiota facilitates the development of immune tolerance. Interventional studies suggest that probiotics could be protective against the development of many diseases. Nevertheless, many factors complicate the analysis of dysbiosis in subjects with food allergy. Comparison in-between studies are difficult, because of considerable heterogeneity in study design, sample size, age at fecal collection, methods of analysis of gut microbiome, and geographic location. Currently, there is no positive recommendation from scientific societies to use pre- or probiotics for treatment of food allergy or other allergic manifestations, while their use in prevention is being custom-cleared. However, the recommendation is still based on little evidence. Although there is valid scientific evidence in vitro, there is no sufficient information to suggest the use of specific probiotics in allergy and asthma prevention.
Collapse
Affiliation(s)
- Maurizio Mennini
- Division of Allergy, Bambino Gesù Children's Hospital, Vatican City, Vatican City
| | - Lamia Dahdah
- Division of Allergy, Bambino Gesù Children's Hospital, Vatican City, Vatican City
| | | | - Alessandro Fiocchi
- Division of Allergy, Bambino Gesù Children's Hospital, Vatican City, Vatican City
| | | |
Collapse
|
49
|
Lactobacillus reuteri DSM 17938 plus vitamin D3 as ancillary treatment in allergic children with asthma. Ann Allergy Asthma Immunol 2016; 117:710-712. [PMID: 27720582 DOI: 10.1016/j.anai.2016.09.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 09/06/2016] [Accepted: 09/07/2016] [Indexed: 11/18/2022]
|
50
|
Dzidic M, Abrahamsson TR, Artacho A, Björkstén B, Collado MC, Mira A, Jenmalm MC. Aberrant IgA responses to the gut microbiota during infancy precede asthma and allergy development. J Allergy Clin Immunol 2016; 139:1017-1025.e14. [PMID: 27531072 DOI: 10.1016/j.jaci.2016.06.047] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 05/11/2016] [Accepted: 06/08/2016] [Indexed: 12/14/2022]
Abstract
BACKGROUND Although a reduced gut microbiota diversity and low mucosal total IgA levels in infancy have been associated with allergy development, IgA responses to the gut microbiota have not yet been studied. OBJECTIVE We sought to determine the proportions of IgA coating together with the characterization of the dominant bacteria, bound to IgA or not, in infant stool samples in relation to allergy development. METHODS A combination of flow cytometric cell sorting and deep sequencing of the 16S rDNA gene was used to characterize the bacterial recognition patterns by IgA in stool samples collected at 1 and 12 months of age from children staying healthy or having allergic symptoms up to 7 years of age. RESULTS The children with allergic manifestations, particularly asthma, during childhood had a lower proportion of IgA bound to fecal bacteria at 12 months of age compared with healthy children. These alterations cannot be attributed to differences in IgA levels or bacterial load between the 2 groups. Moreover, the bacterial targets of early IgA responses (including coating of the Bacteroides genus), as well as IgA recognition patterns, differed between healthy children and children with allergic manifestations. Altered IgA recognition patterns in children with allergy were observed already at 1 month of age, when the IgA antibodies are predominantly maternally derived in breast-fed children. CONCLUSION An aberrant IgA responsiveness to the gut microbiota during infancy precedes asthma and allergy development, possibly indicating an impaired mucosal barrier function in allergic children.
Collapse
Affiliation(s)
- Majda Dzidic
- Department of Clinical and Experimental Medicine, Unit of Autoimmunity and Immune Regulation, Linköping University, Linköping, Sweden; Department of Health and Genomics, FISABIO Foundation, Center for Advanced Research in Public Health, Valencia, Spain; Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Department of Biotechnology, Unit of Lactic Acid Bacteria and Probiotics, Valencia, Spain
| | - Thomas R Abrahamsson
- Department of Clinical and Experimental Medicine, Division of Paediatrics, Linköping University, Linköping, Sweden
| | - Alejandro Artacho
- Department of Health and Genomics, FISABIO Foundation, Center for Advanced Research in Public Health, Valencia, Spain
| | - Bengt Björkstén
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Department of Biotechnology, Unit of Lactic Acid Bacteria and Probiotics, Valencia, Spain
| | - Alex Mira
- Department of Health and Genomics, FISABIO Foundation, Center for Advanced Research in Public Health, Valencia, Spain.
| | - Maria C Jenmalm
- Department of Clinical and Experimental Medicine, Unit of Autoimmunity and Immune Regulation, Linköping University, Linköping, Sweden.
| |
Collapse
|