1
|
Liang C, Zhang LW. Profiling the Gut Microbiota in Obese Children with Formula Feeding in Early Life and Selecting Strains against Obesity. Foods 2024; 13:1379. [PMID: 38731751 PMCID: PMC11083066 DOI: 10.3390/foods13091379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/23/2024] [Accepted: 04/27/2024] [Indexed: 05/13/2024] Open
Abstract
Formula feeding, obesity and the gut microbiota are closely related. The present investigation explored the profiles of the intestinal microbiota in obese children over 5 years old with formula feeding in early life. We identified functional bacteria with anti-obesity potential through in vitro and in vivo experiments, elucidating their mechanisms. The results indicated that, in the group of children over 5 years old who were fed formula in early life, obese children exhibited distinct gut microbiota, which were characterized by diminished species diversity and reduced Bifidobacterium levels compared to normal-weight children. As a result, Lactobacillus acidophilus H-68 (H-68) was isolated from the feces of the N-FF group and recognized as a promising candidate. H-68 demonstrated the ability to stimulate cholecystokinin (CCK) secretion in STC-1 cells and produce bile salt hydrolase. In vivo, H-68 promoted CCK secretion, suppressing food intake, and regulated bile acid enterohepatic circulation, leading to increased deoxycholic acid and lithocholic acid levels in the ileum and liver. This regulation effectively inhibited the diet-induced body weight and body fat gain, along with the liver fat deposition. In conclusion, H-68 was recognized for its prospective anti-obesity impact, signifying an auspicious pathway for forthcoming interventions targeted at averting pediatric obesity in formula-fed children.
Collapse
Affiliation(s)
- Cong Liang
- College of Safety and Environmental Engineering, Shandong University of Science and Technology, Qingdao 266590, China;
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150010, China
| | - Lan-Wei Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
| |
Collapse
|
2
|
Montoya CA, Young W, Ryan L, Dunstan K, Peters J, Dewhurst H, Dekker J, Haggarty N, Dilger RN, Roy NC. The probiotic Lacticaseibacillus rhamnosus HN001 influences the architecture and gene expression of small intestine tissue in a piglet model. Br J Nutr 2024; 131:1289-1297. [PMID: 38053344 PMCID: PMC10950449 DOI: 10.1017/s0007114523002830] [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: 04/17/2023] [Revised: 10/25/2023] [Accepted: 11/24/2023] [Indexed: 12/07/2023]
Abstract
This study investigated the effects of Lacticaseibacillus rhamnosus HN001 supplementation on the architecture and gene expression in small intestinal tissues of piglets used as an animal model for infant humans. Twenty-four 10-d-old entire male piglets (4·3 (sd 0·59) kg body weight) were fed an infant formula (IF) (control) or IF supplemented with 1·3 × 105 (low dose) or 7·9 × 106 (high dose) colony-forming units HN001 per ml of reconstituted formula (n 8 piglets/treatment). After 24 d, piglets were euthanised. Samples were collected to analyse the histology and gene expression (RNAseq and qPCR) in the jejunal and ileal tissues, blood cytokine concentrations, and blood and faecal calprotectin concentrations. HN001 consumption altered (false discovery rate < 0·05) gene expression (RNAseq) in jejunal tissues but not in ileal tissues. The number of ileal goblet cells and crypt surface area increased quadratically (P < 0·05) as dietary HN001 levels increased, but no increase was observed in the jejunal tissues. Similarly, blood plasma concentrations of IL-10 and calprotectin increased linearly (P < 0·05) as dietary HN001 levels increased. In conclusion, supplementation of IF with HN001 affected the architecture and gene expression of small intestine tissue, blood cytokine concentration and frequencies, and blood calprotectin concentrations, indicating that HN001 modulated small intestinal tissue maturation and immunity in the piglet model.
Collapse
Affiliation(s)
- Carlos A. Montoya
- Smart Foods & Bioproducts, AgResearch, Te Ohu Rangahau Kai Facility, Palmerston North, New Zealand
- Riddet Institute, Massey University, Te Ohu Rangahau Kai Facility, Palmerston North4474, New Zealand
| | - Wayne Young
- Smart Foods & Bioproducts, AgResearch, Te Ohu Rangahau Kai Facility, Palmerston North, New Zealand
- Riddet Institute, Massey University, Te Ohu Rangahau Kai Facility, Palmerston North4474, New Zealand
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
- Fonterra Research and Development Centre, Dairy Farm Rd, Palmerston North, New Zealand
| | - Leigh Ryan
- Smart Foods & Bioproducts, AgResearch, Te Ohu Rangahau Kai Facility, Palmerston North, New Zealand
| | - Kelly Dunstan
- Smart Foods & Bioproducts, AgResearch, Te Ohu Rangahau Kai Facility, Palmerston North, New Zealand
| | - Jason Peters
- Smart Foods & Bioproducts, AgResearch, Te Ohu Rangahau Kai Facility, Palmerston North, New Zealand
| | - Hilary Dewhurst
- Smart Foods & Bioproducts, AgResearch, Te Ohu Rangahau Kai Facility, Palmerston North, New Zealand
| | - James Dekker
- Fonterra Research and Development Centre, Dairy Farm Rd, Palmerston North, New Zealand
| | - Neill Haggarty
- Fonterra Research and Development Centre, Dairy Farm Rd, Palmerston North, New Zealand
| | - Ryan N. Dilger
- Department of Animal Sciences, University of Illinois, Urbana, IL, USA
| | - Nicole C. Roy
- Riddet Institute, Massey University, Te Ohu Rangahau Kai Facility, Palmerston North4474, New Zealand
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
- Department of Human Nutrition, University of Otago, Dunedin, New Zealand
| |
Collapse
|
3
|
Yu P, Pan Y, Pei Z, Guo M, Yang B, Lee YK, Liu X, Zhao J, Zhang H, Chen W. Influence of Lactose Supplementation on Regulation of Streptococcus thermophilus on Gut Microbiota. Nutrients 2023; 15:4767. [PMID: 38004159 PMCID: PMC10675825 DOI: 10.3390/nu15224767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
It has been found that Streptococcus thermophilus (S. thermophilus) influenced the gut microbiota and host metabolism with strain specificity in C57BL/6J mice in the previous study, though it remains unclear whether lactose as a dietary factor associated with dairy consumption is involved as the mediator in the interaction. In the present study, integrated analysis of 16S rRNA gene sequencing and untargeted metabolomics by liquid chromatography-mass spectrometry of fecal samples in C57BL/6J mice was applied to evaluate the effect of lactose on the regulation of gut microbiota by two S. thermophilus strains (4M6 and DYNDL13-4). The results showed that the influence of lactose supplementation on gut microbiota induced by S. thermophilus ingestion was strain-specific. Although two S. thermophilus strains ingestion introduced similar perturbations in the fecal microbiota and gut microbial metabolism, the regulation of DYNDL13-4 on the gut microbiota and metabolism was more affected by lactose than 4M6. More specifically, lactose and 4M6 supplementation mainly enriched pathways of d-glutamine and d-glutamate metabolism, alanine, aspartate, and glutamate metabolism, and tryptophan and phenylalanine metabolism in the gut, whereas 4M6 only enriched tryptophan and phenylalanine metabolism. DYNDL13-4-L (DYNDL13-4 with lactose) had significant effects on sulfur, taurine, and hypotaurine metabolism in the gut and on phenylalanine, tyrosine, tryptophan biosynthesis, and linoleic acid metabolism in serum relative to the DYNDL13-4. Our study demonstrated the strain-specific effect of lactose and S. thermophilus supplementation on gut microbiota and host metabolism. However, considering the complexity of the gut microbiota, further research is necessary to provide insights to facilitate the design of personalized fermented milk products as a dietary therapeutic strategy for improving host health.
Collapse
Affiliation(s)
- Peng Yu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China (Z.P.); (B.Y.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China;
| | - Yuqi Pan
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China (Z.P.); (B.Y.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China;
| | - Zhiwen Pei
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China (Z.P.); (B.Y.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China;
| | - Min Guo
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China;
| | - Bo Yang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China (Z.P.); (B.Y.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China;
- International Joint Research Laboratory for Pharmabiotics & Antibiotic Resistance, Jiangnan University, Wuxi 214122, China;
| | - Yuan-Kun Lee
- International Joint Research Laboratory for Pharmabiotics & Antibiotic Resistance, Jiangnan University, Wuxi 214122, China;
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Xiaoming Liu
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China;
- International Joint Research Laboratory for Pharmabiotics & Antibiotic Resistance, Jiangnan University, Wuxi 214122, China;
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China (Z.P.); (B.Y.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China;
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China (Z.P.); (B.Y.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China;
- Wuxi Translational Medicine Research Center, Wuxi 214122, China
- Jiangsu Translational Medicine Research Institute, Wuxi Branch, Wuxi 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China (Z.P.); (B.Y.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China;
- National Engineering Research Centre for Functional Food, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
4
|
Khablenko A, Danylenko S, Yalovenko O, Duhan O, Potemskaia O, Prykhodko D. Recombinant Probiotic Preparations: Current State, Development and Application Prospects. INNOVATIVE BIOSYSTEMS AND BIOENGINEERING 2023; 6:119-147. [DOI: 10.20535/ibb.2022.6.3-4.268349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
The article is devoted to the latest achievements in the field of research, development, and implementation of various types of medicinal products based on recombinant probiotics. The benefits of probiotics, their modern use in medicine along with the most frequently used genera and species of probiotic microorganisms were highlighted. The medicinal and therapeutic activities of the studied probiotics were indicated. The review suggests various methods of creating recombinant probiotic microorganisms, including standard genetic engineering methods, as well as systems biology approaches and new methods of using the CRISPR-Cas system. The range of potential therapeutic applications of drugs based on recombinant probiotics was proposed. Special attention was paid to modern research on the creation of new, more effective recombinant probiotics that can be used for various therapeutic purposes. Considering the vast diversity of therapeutic applications of recombinant probiotics and ambiguous functions, their use for the potential treatment of various common human diseases (non-infectious and infectious diseases of the gastrointestinal tract, metabolic disorders, and allergic conditions) was investigated. The prospects for creating different types of vaccines based on recombinant probiotics together with the prospects for their implementation into medicine were considered. The possibilities of using recombinant probiotics in veterinary medicine, particularly for the prevention of domestic animal diseases, were reviewed. The prospects for the implementation of recombinant probiotics as vaccines and diagnostic tools for testing certain diseases as well as modeling the work of the human digestive system were highlighted. The risks of creation, application, including the issues related to the regulatory sphere regarding the use of new recombinant microorganisms, which can potentially enter the environment and cause unforeseen circumstances, were outlined.
Collapse
Affiliation(s)
| | - Svetlana Danylenko
- Institute of Food Resources of the National Academy of Agrarian Sciences of Ukraine, Ukraine
| | | | - Olexii Duhan
- Igor Sikorsky Kyiv Polytechnic Institute, Ukraine
| | - Oksana Potemskaia
- Institute of Food Resources of the National Academy of Agrarian Sciences of Ukraine, Ukraine
| | | |
Collapse
|
5
|
Indrio F, Gutierrez Castrellon P, Vandenplas Y, Cagri Dinleyici E, Francavilla R, Mantovani MP, Grillo A, Beghetti I, Corvaglia L, Aceti A. Health Effects of Infant Formula Supplemented with Probiotics or Synbiotics in Infants and Toddlers: Systematic Review with Network Meta-Analysis. Nutrients 2022; 14:5175. [PMID: 36501205 PMCID: PMC9739048 DOI: 10.3390/nu14235175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 11/27/2022] [Accepted: 12/02/2022] [Indexed: 12/09/2022] Open
Abstract
Supplementation of infant and follow-up formula with probiotics or synbiotics has become a common practice. In 2011 and 2017, the evidence regarding the impact of these interventions was analysed systematically. Recently new evidence was published. To evaluate through a systematic review with network meta-analysis the evidence on the impact of infant formula supplemented with probiotics or synbiotics for healthy infants and 36-month-old toddlers. RCTs published between 1999-2019 for infant formulas supplemented with probiotics alone or synbiotics in healthy infants and toddlers were identified. Data analysis included clinical (gastrointestinal symptoms, risk reduction of infectious diseases, use of antibiotics, weight/height gain and frequency of adverse events) and non-clinical outcomes (changes in faecal microbiota and immune parameters). A random effect model was used. Hedges' standard mean difference (SMD) and risk ratio (RR) were calculated. Rank analysis was performed to evaluate the superiority of each intervention. Twenty-six randomised controlled trials with 35 direct comparisons involving 1957 children receiving probiotic-supplemented formula and 1898 receiving control formula were reviewed. The mean duration of intervention was 5.6 ± 2.84 months. Certain strains demonstrated a reduction in episodes of colic, number of days with fever and use of antibiotics; however, there was considerable heterogeneity which reduced the level of certainty of effect. No significant effects were observed on weight, height or changes in faecal proportions of Bifidobacteria, Lactobacillus, Bacteroides or Clostridia. Although there is some evidence that may support a potential benefit of probiotic or synbiotic supplementation of infant formulas, variation in the quality of existing trials and the heterogeneity of the data preclude the establishment of robust recommendations.
Collapse
Affiliation(s)
- Flavia Indrio
- Department of Medical and Surgical Science Pediatric Section, University of Foggia, 71100 Foggia, Italy
| | - Pedro Gutierrez Castrellon
- Centro de Investigación Translacional en Ciencias de la Salud, Hospital General Dr. Manuel Gea González, 14080 Ciudad de México, Mexico
| | - Yvan Vandenplas
- UZ Brussel, KidZ Health Castle, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Ener Cagri Dinleyici
- Department of Pediatrics, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir 26480, Turkey
| | - Ruggiero Francavilla
- Interdisciplinary Department of Medicine, Pediatric Section, Children’s Hospital ‘Giovanni XXIII’, University of Bari Aldo Moro, 70126 Bari, Italy
| | | | - Assunta Grillo
- Department of Medical and Surgical Science Pediatric Section, University of Foggia, 71100 Foggia, Italy
| | - Isadora Beghetti
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
- Neonatal Intensive Care Unit, IRCCS AOUBO, 40138 Bologna, Italy
| | - Luigi Corvaglia
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
- Neonatal Intensive Care Unit, IRCCS AOUBO, 40138 Bologna, Italy
| | - Arianna Aceti
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
- Neonatal Intensive Care Unit, IRCCS AOUBO, 40138 Bologna, Italy
| |
Collapse
|
6
|
Hubbard GP, Atwal K, Graham L, Narayanan S, Cooke L, Casewell C, Denton SA, Gavin J, Browne RM, Kinnear FJ, McHardy AJ, Evans D, Vallis R, Venkataraman D, Cawood AL, Donohoe S, Steele V, Armstrong S, Stratton RJ. Synbiotic containing extensively hydrolyzed formula improves gastrointestinal and atopic symptom severity, growth, caregiver quality of life, and hospital-related healthcare use in infants with cow's milk allergy. Immun Inflamm Dis 2022; 10:e636. [PMID: 35634950 PMCID: PMC9119008 DOI: 10.1002/iid3.636] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 04/28/2022] [Accepted: 05/01/2022] [Indexed: 12/13/2022] Open
Abstract
Background Healthy gut microbiota is important for prognosis in cow's milk allergy (CMA). The application of synbiotics (specific pre‐ and probiotics) in extensively hydrolyzed formulae (eHFs) is a relatively new concept. Aims To evaluate a synbiotic‐containing, whey‐based eHF (SeHF) with galacto‐oligosaccharides, fructo‐oligosaccharides, and bifidobacterium breve M‐16V in infants with CMA. Materials and Methods A 31‐day one‐arm pilot study in 29 infants with CMA (mean age 30.8 weeks [SD 11]) was undertaken, with outcomes including gastrointestinal tolerance, atopic dermatitis symptoms, dietary intake, growth, SeHF acceptability, caregiver quality of life, and hospital‐related healthcare use. Results Significant improvements (p < .05) in the severity of abdominal pain (in 57%), burping (in 46%), flatulence (in 79%), constipation (in 14%), rhinitis (41%), and itchy eyes (73%), as well as atopic dermatitis in those with severe baseline symptoms (PO‐SCORAD© reduction: 34.7–18.2 (p = .003), n = 6) were observed over time. Growth and caregiver quality of life scores significantly increased (+26.7%, p < .05) over time. Hospital visits and medications significantly reduced (−1.61 and −2.23, respectively, p < .005) in the 6 months after SeHF initiation. Discussion In this small, single‐arm, pilot study, the use of SeHF enhanced the management of infants with non‐IgE mediated CMA who were already established on eHF. Conclusion: Whilst this study adds to the evidence base for the use of SeHF in CMA, further robust research to explore the longer‐term benefits of synbiotics, specifically the blend used in this study, for the clinical management of infants with CMA is warranted.
Collapse
Affiliation(s)
| | | | - Lynne Graham
- West Hertfordshire Hospitals NHS Trust, Watford, UK
| | | | - Lisa Cooke
- Bristol Royal Hospital for Children, Bristol, UK
| | | | - Sally-Ann Denton
- University Hospitals Southampton NHS Foundation Trust, Southampton, UK
| | - Joan Gavin
- University Hospitals Southampton NHS Foundation Trust, Southampton, UK
| | | | | | - Ailsa J McHardy
- University Hospitals Southampton NHS Foundation Trust, Southampton, UK
| | - Debbie Evans
- Royal Surrey NHS Foundation Trust, Guildford, UK
| | | | | | | | - Sarah Donohoe
- James Cook Hospital, South Tees Hospitals NHS Foundation Trust, Middlesbrough, UK
| | - Victoria Steele
- James Cook Hospital, South Tees Hospitals NHS Foundation Trust, Middlesbrough, UK
| | - Sonia Armstrong
- James Cook Hospital, South Tees Hospitals NHS Foundation Trust, Middlesbrough, UK
| | - Rebecca J Stratton
- Nutricia Ltd, Trowbridge, UK.,Faculty of Medicine, University of Southampton, Southampton, UK
| |
Collapse
|
7
|
Aziz G, Zaidi A, Tariq M. Compositional Quality and Possible Gastrointestinal Performance of Marketed Probiotic Supplements. Probiotics Antimicrob Proteins 2022; 14:288-312. [PMID: 35199309 DOI: 10.1007/s12602-022-09931-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2022] [Indexed: 12/15/2022]
Abstract
The local pharmacies and shops are brimming with various probiotic products that herald a range of health benefits. The poor quality of probiotic products in both dosage and species is symptomatic of this multi-billion-dollar market making it difficult for consumers to single out reliable ones. This study aims to fill the potential gap in the labeling accuracy of probiotic products intended for human consumption. We describe a combinatorial approach using classical culture-dependent technique to quantify and molecular techniques (16 s rRNA gene sequencing, multilocus sequence, and ribotyping) for strain recognition of the microbial contents. The full gamut of probiotic characteristics including acid, bile and lysozyme tolerances, adhesiveness, anti-pathogenicity, and degree of safeness were performed. Their capacity to endure gastro-intestinal (GIT) stresses and select drugs was assessed in vitro. Our results forced us to declare that the local probiotic market is essentially unregulated. Almost none of the probiotic products tested met the label claim. Some (11%) have no viable cells, and a quarter (27%) showing significant inter-batch variation. A lower microbial count was typical with undesirables constituting a quarter of the total (~ 27%). Half of the products contained antibiotic-resistant strains; the unregulated use of these probiotics carries the risk of spreading antibiotic resistance to gut pathobionts. Poor tolerance to gut conditions and mediocre functionalism make the case worse. The current regulatory systems do not take this discrepancy into account. We recommend an evidence-based regular market surveillance of marketed probiotics to ensure the authenticity of the claims and product effectiveness.
Collapse
Affiliation(s)
- Ghazal Aziz
- National Probiotic Laboratory, National Institute for Biotechnology and Genetic Engineering College (NIBGE-C)-PIEAS, Faisalabad, 38000, Punjab, Pakistan
- Pakistan Institute of Engineering and Applied Sciences (PIEAS), Nilore, Islamabad, 45650, ICT, Pakistan
| | - Arsalan Zaidi
- National Probiotic Laboratory, National Institute for Biotechnology and Genetic Engineering College (NIBGE-C)-PIEAS, Faisalabad, 38000, Punjab, Pakistan.
- Pakistan Institute of Engineering and Applied Sciences (PIEAS), Nilore, Islamabad, 45650, ICT, Pakistan.
| | - Muhammad Tariq
- National Probiotic Laboratory, National Institute for Biotechnology and Genetic Engineering College (NIBGE-C)-PIEAS, Faisalabad, 38000, Punjab, Pakistan
- Pakistan Institute of Engineering and Applied Sciences (PIEAS), Nilore, Islamabad, 45650, ICT, Pakistan
| |
Collapse
|
8
|
Penhasi A, Baluashvili I, Shalev DE. An edible erodible semi‐interpenetrating polymer network as a microcapsule shell to protect probiotic bacteria during reconstitution of powdered infant formula at elevated temperatures. J Appl Polym Sci 2022. [DOI: 10.1002/app.52201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Adel Penhasi
- Department of Research and Development PolyCaps Ltd. Tel Aviv Israel
- Department of Pharmaceutical Engineering Azrieli College of Engineering Jerusalem Jerusalem Israel
| | | | - Deborah E. Shalev
- Department of Pharmaceutical Engineering Azrieli College of Engineering Jerusalem Jerusalem Israel
- Wolfson Centre for Applied Structural Biology The Hebrew University of Jerusalem, Edmond J. Safra Campus Jerusalem Israel
| |
Collapse
|
9
|
Michels M, Córneo E, Cucker L, Simon CS, de Jesus GFA, Michels C, Madeira K, Voytena AP, Rossetto M, Ramlov F, de Castro Damásio D, Dal-Pizzol F. Bifidobacterium lactis CCT 7858 Improves Gastrointestinal Symptoms by Antibiotics Treatment: a Double-Blind, Randomized, Placebo-Controlled Trial. Probiotics Antimicrob Proteins 2022; 15:738-748. [PMID: 35031969 DOI: 10.1007/s12602-021-09900-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/24/2021] [Indexed: 01/30/2023]
Abstract
Diarrhea is one of the most frequent side effects of antibiotic treatment and occurs in 25 to 40% of patients in use. One potential strategy to prevent this side effect is the concurrent use of probiotics. This study evaluated the efficacy of the strain Bifidobacterium lactis CCT 7858 in the prevention of diarrhea and improvement of gastrointestinal symptoms in hospitalized patients using antibiotics. This was a randomized, blinded, placebo-controlled clinical trial. This study included 104 patients in antibiotic treatment. Patients were randomized into two groups: placebo (maltodextrin) and intervention (strain Bifidobacterium lactis CCT 7858 at 9 × 1010 CFU concentration; GABBIA® Biotecnology, Santa Catarina, Brazil). Patients were supplemented depending on the duration of antibiotic therapy, and both were evaluated with scales in two moments: before and after treatment. We included 104 hospitalized patients. In follow-up, 38 (74.5%) of the B. lactis group have no reported diarrhea. In secondary outcomes, in five day strong abdominal distension was reported in 4 (7,3) placebo group and not reported in B. lactis. Abdominal noises, nausea, and vomiting were not registered in any group. B. lactis strain has been considered safe and with several benefits, including reduction of soft stools and gastrointestinal symptoms how abdominal noise, pain and distension, as well reduction of diarrhea.
Collapse
Affiliation(s)
- Monique Michels
- Gabbia Biotechnology, Barra Velha, Santa Catarina, Brazil.,Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Avenida Universitária, Bairro Universitário, Criciúma, SC, 1105, Brazil
| | - Emily Córneo
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Avenida Universitária, Bairro Universitário, Criciúma, SC, 1105, Brazil
| | - Luana Cucker
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Avenida Universitária, Bairro Universitário, Criciúma, SC, 1105, Brazil
| | - Carla Sasso Simon
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Avenida Universitária, Bairro Universitário, Criciúma, SC, 1105, Brazil
| | | | - Carolina Michels
- Laboratory of Quantitative Methods, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Kristian Madeira
- Laboratory of Quantitative Methods, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | | | | | | | - Danusa de Castro Damásio
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Avenida Universitária, Bairro Universitário, Criciúma, SC, 1105, Brazil.,São José Hospital Research Centre, Rua Coronel Pedro Benedet, Criciúma, SC, 63088801-460, Brazil
| | - Felipe Dal-Pizzol
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Avenida Universitária, Bairro Universitário, Criciúma, SC, 1105, Brazil. .,São José Hospital Research Centre, Rua Coronel Pedro Benedet, Criciúma, SC, 63088801-460, Brazil.
| |
Collapse
|
10
|
Catania J, Pandit NG, Ehrlich JM, Zaman M, Stone E, Franceschi C, Smith A, Tanner-Smith E, Zackular JP, Bhutta ZA, Imdad A. Probiotic Supplementation for Promotion of Growth in Children: A Systematic Review and Meta-Analysis. Nutrients 2021; 14:83. [PMID: 35010959 PMCID: PMC8746675 DOI: 10.3390/nu14010083] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/18/2021] [Accepted: 12/22/2021] [Indexed: 11/16/2022] Open
Abstract
Probiotics are commonly prescribed to promote a healthy gut microbiome in children. Our objective was to investigate the effects of probiotic supplementation on growth outcomes in children 0-59 months of age. We conducted a systematic review and meta-analysis which included randomized controlled trials (RCTs) that administered probiotics to children aged 0-59 months, with growth outcomes as a result. We completed a random-effects meta-analysis and calculated a pooled standardized mean difference (SMD) or relative risk (RR) and reported with a 95% confidence interval (CI). We included 79 RCTs, 54 from high-income countries (HIC), and 25 from low- and middle-income countries (LMIC). LMIC data showed that probiotics may have a small effect on weight (SMD: 0.26, 95% CI: 0.11-0.42, grade-certainty = low) and height (SMD 0.16, 95% CI: 0.06-0.25, grade-certainty = moderate). HIC data did not show any clinically meaningful effect on weight (SMD: 0.01, 95% CI: -0.04-0.05, grade-certainty = moderate), or height (SMD: -0.01, 95% CI: -0.06-0.04, grade-certainty = moderate). There was no evidence that probiotics affected the risk of adverse events. We conclude that in otherwise healthy children aged 0-59 months, probiotics may have a small but heterogenous effect on weight and height in LMIC but not in children from HIC.
Collapse
Affiliation(s)
- Joseph Catania
- College of Medicine, SUNY Upstate Medical University, Syracuse, NY 13210, USA; (J.C.); (N.G.P.); (J.M.E.); (M.Z.); (E.S.); (C.F.)
| | - Natasha G. Pandit
- College of Medicine, SUNY Upstate Medical University, Syracuse, NY 13210, USA; (J.C.); (N.G.P.); (J.M.E.); (M.Z.); (E.S.); (C.F.)
| | - Julie M. Ehrlich
- College of Medicine, SUNY Upstate Medical University, Syracuse, NY 13210, USA; (J.C.); (N.G.P.); (J.M.E.); (M.Z.); (E.S.); (C.F.)
| | - Muizz Zaman
- College of Medicine, SUNY Upstate Medical University, Syracuse, NY 13210, USA; (J.C.); (N.G.P.); (J.M.E.); (M.Z.); (E.S.); (C.F.)
| | - Elizabeth Stone
- College of Medicine, SUNY Upstate Medical University, Syracuse, NY 13210, USA; (J.C.); (N.G.P.); (J.M.E.); (M.Z.); (E.S.); (C.F.)
| | - Courtney Franceschi
- College of Medicine, SUNY Upstate Medical University, Syracuse, NY 13210, USA; (J.C.); (N.G.P.); (J.M.E.); (M.Z.); (E.S.); (C.F.)
| | - Abigail Smith
- Health Science Library, SUNY Upstate Medical University, Syracuse, NY 13210, USA;
| | | | - Joseph P. Zackular
- Department of Pathology, Children’s Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Zulfiqar A. Bhutta
- Centre for Global Child Health, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada;
- Center of Excellence in Women and Child Health, The Aga Khan University, Karachi 74800, Pakistan
| | - Aamer Imdad
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology and Nutrition, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| |
Collapse
|
11
|
Ahmad HH, Peck B, Terry D. The influence of probiotics on gastrointestinal tract infections among children attending childcare: A systematic review and meta-analysis. J Appl Microbiol 2021; 132:1636-1651. [PMID: 34796583 DOI: 10.1111/jam.15374] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/07/2021] [Accepted: 11/17/2021] [Indexed: 12/11/2022]
Abstract
Current literature related to the impact of probiotics on the incidence of gastrointestinal tract infections (GITIs) has shown mixed results and no systematic review available with pooled analysis exists. Thus, the aim of this systematic review was to provide contemporary evidence regarding the overall and strain-specific influence of probiotics in preventing GITIs among infants and children attending childcare centres. The review shortlisted 18 RCTs after screening through the initial search results of 779 articles. However, only 15 trials were deemed eligible, addressing at least one outcome in the pooled analysis. It is concluded that the supplementation of probiotics (overall effect) may reduce the risk of GITI episode by 26%, with Lacticaseibacillus paracasei, Limosilactobacillus reuteri and Lacticaseibacillus rhamnosus GG being specifically potent probiotic strains in reducing GITI episode, duration of infection and absence from childcare respectively. There is insufficient evidence to determine the effect of Bifidobacterium animalis subsp. lactis BB-12 based on the findings of the trials included in this review.
Collapse
Affiliation(s)
- Hafiz H Ahmad
- School of Health, Federation University, Ballarat, Victoria, Australia
| | - Blake Peck
- School of Health, Federation University, Ballarat, Victoria, Australia
| | - Daniel Terry
- School of Health, Federation University, Ballarat, Victoria, Australia
| |
Collapse
|
12
|
Piątek J, Bernatek M, Krauss H, Wojciechowska M, Chęcińska-Maciejewska Z, Kaczmarek P, Sommermeyer H. Effects of a nine-strain bacterial synbiotic compared to simethicone in colicky babies - an open-label randomised study. Benef Microbes 2021; 12:249-257. [PMID: 33765904 DOI: 10.3920/bm2020.0160] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The aim of the study was to determine effects of administration of simethicone and a multi-strain synbiotic on the crying behaviour of colicky babies. The study design consisted of an open-label, two parallel treatment group study involving 87 infants aged 3-6 weeks with infantile colic (defined as crying episodes lasting 3 or more hours per day and occurring at least 3 days per week within 3 weeks prior to enrolment) randomly, unequally [1:1.5] assigned to receive simethicone (n=33) or a multi-strain synbiotic (n=54) orally for 4 weeks. The multi-strain synbiotic contained Lactobacillus acidophilus LA-14, Lacticaseibacillus casei R0215, Lacticaseibacillus paracasei Lp-115, Lacticaseibacillus rhamnosus GG, Ligilactobacillus salivarius Ls-33, Bifidobacterium lactis Bl-04, Bifidobacterium bifidum R0071, Bifidobacterium longum R0175 and fructooligosaccharides). Primary outcome measures were the responder rates (effect ≥50% reduction from baseline) of the measures 'crying days last 3 weeks', 'average evening crying duration last 3 weeks' and 'reduction of average number of crying phases per day last three weeks' at the end of treatment. The study is registered at ClinicalTrials.gov under NCT04487834. Significantly higher responder rates (effect ≥50% reduction from baseline) of the multi-strain synbiotic compared to simethicone were found for the measures 'crying days last 3 weeks' (72% vs 18%, P<0.0001) and 'average evening crying duration last 3 weeks' (85% vs 39%, P=0.0001). No significant difference was found for the measure 'reduction of average number of crying phases per day last three weeks' (50% vs 42%, P=0.4852). No adverse effects were reported for the two treatment groups. Based on these results, the multi-strain synbiotic can be considered as an interesting therapeutic possibility for the treatment of infantile colic, worthwhile to be investigated further in non-clinical and clinical studies.
Collapse
Affiliation(s)
- J Piątek
- Department of Medicine, The President Stanisław Wojciechowski State University of Applied Sciences in Kalisz, Nowy Šwiat 4, 62-800 Kalisz, Poland
| | - M Bernatek
- Department of Medicine, The President Stanisław Wojciechowski State University of Applied Sciences in Kalisz, Nowy Šwiat 4, 62-800 Kalisz, Poland
| | - H Krauss
- Department of Medicine, The President Stanisław Wojciechowski State University of Applied Sciences in Kalisz, Nowy Šwiat 4, 62-800 Kalisz, Poland
| | - M Wojciechowska
- Mother and Child Health Department, Karol Marcinkowski Medical University in Poznań, ul. Fredry 10, 61-701 Poznan, Poland
| | - Z Chęcińska-Maciejewska
- Physiology Department, Karol Marcinkowski Medical University in Poznań, ul. Fredry 10, 61-701 Poznan, Poland
| | - P Kaczmarek
- Department of Medicine, The President Stanisław Wojciechowski State University of Applied Sciences in Kalisz, Nowy Šwiat 4, 62-800 Kalisz, Poland
| | - H Sommermeyer
- Department of Medicine, The President Stanisław Wojciechowski State University of Applied Sciences in Kalisz, Nowy Šwiat 4, 62-800 Kalisz, Poland
| |
Collapse
|
13
|
Chen K, Liu C, Li H, Lei Y, Zeng C, Xu S, Li J, Savino F. Infantile Colic Treated With Bifidobacterium longum CECT7894 and Pediococcus pentosaceus CECT8330: A Randomized, Double-Blind, Placebo-Controlled Trial. Front Pediatr 2021; 9:635176. [PMID: 34568236 PMCID: PMC8461252 DOI: 10.3389/fped.2021.635176] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 08/12/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Colic is a common condition in infants <4 months of age. Attempts to treat infantile colic with probiotics have shown variable efficacy and overall low evidence of success. In this work, we tested the hypothesis that oral administration of Bifidobacterium longum CECT7894 (KABP042) and Pediococcus pentosaceus CECT8330 (KABP041) mix (1 × 109 colony forming units) would improve the symptoms of infantile colic. Methods: A total of 112 exclusively breastfed or mixed fed infants aged <2 months and meeting the ROME IV criteria for infantile colic were recruited. The infants were randomized in a double-blind, placebo-controlled trial to receive orally administered probiotics (intervention group, IG, n = 48) or placebo (placebo group, PG, n = 42) daily for 21 days. Results: Infants in the IG had significantly shorter crying time (p < 0.001) on day 7 [IG vs. PG, median (25-75th percentile): 38 (3.5-40.5) vs. 62 (40-108) min/day], day 14 [IG vs. PG: 20 (0-40) vs. 50 (30-75) min/day], and day 21 [IG vs. PG: 14 (0-33) vs. 40 (28-62) min/day]. Higher responder ratio and fewer crying/fussing episodes on days 7, 14, and 21 and better stool consistency on day 21 were observed in the IG (p < 0.01) as compared to the PG. Conversely, no significant effects on stool frequency or quality of life were observed. Conclusions: In summary, daily oral administration of B. longum CECT7894 (KABP042) and P. pentosaceus CECT8330 (KABP041) was an effective treatment for shortening crying time due to infantile colic and for improving fecal consistency. This trial was registered retrospectively in December 2019 with a trial number of ISRCTN92431452.
Collapse
Affiliation(s)
- Ke Chen
- Department of Nutrition, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.,Department of Child Health Care, Angel Children's Hospital, Chengdu, China
| | - Changqi Liu
- School of Exercise and Nutritional Sciences, San Diego State University, San Diego, CA, United States
| | - Hua Li
- Department of Child Health Care, Qingbaijiang Maternal and Child Health Hospital, Chengdu, China
| | - Yuehua Lei
- Department of Child Health Care, Qingbaijiang Maternal and Child Health Hospital, Chengdu, China
| | - Chenggui Zeng
- Department of Child Health Care, Chengdu Caojiaxiang Community Healthcare Center, Chengdu, China
| | - Shuhong Xu
- Department of Child Health Care, Huili Maternity and Child Care Center, Huili, China
| | - Jianqiu Li
- Department of Child Health Care, Angel Children's Hospital, Chengdu, China
| | - Francesco Savino
- Department of Paediatrics, S.S.D. Subintensive Neonatal Care, Children Hospital 'Regina Margherita', Turin, Italy
| |
Collapse
|
14
|
Ouald Chaib A, Levy EI, Ouald Chaib M, Vandenplas Y. The influence of the gastrointestinal microbiome on infant colic. Expert Rev Gastroenterol Hepatol 2020; 14:919-932. [PMID: 32633578 DOI: 10.1080/17474124.2020.1791702] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Although infantile colic is relatively frequent, its pathophysiology is not yet understood. The aim of this paper is to provide a better understanding of the link between infantile colic and the gastrointestinal microbiome. AREAS COVERED The gastro-intestinal microbiome may already start to develop in the womb and grows exponentially immediately after birth. Factors influencing the microbiome can cause dysbiosis and precipitate symptoms of colic through several mechanisms such as increased gas production and low grade gut inflammation. Other possible factors are immaturity of the enterohepatic bile acid cycle and administration of antibiotics and other medications during the perinatal period. An effective treatment for all colicky infants has yet to be discovered, but the probiotic Lactobacillus reuteri DSM17938 was shown to be effective in breastfed infants with colic. The scientific databases 'Pubmed' and 'Google scholar' were searched from inception until 02/2020. Relevant articles were selected based on the abstract. EXPERT OPINION Recent literature confirmed that the composition of the gastrointestinal microbiome is associated with the development of infantile colic. It can be speculated that full sequencing and bioinformatics analysis to identify the microbiome down to the species level may provide answers to the etiology and management of infantile colic.
Collapse
Affiliation(s)
- Abdelhalim Ouald Chaib
- KidZ Health Castle, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel , Brussels, Belgium
| | - Elvira Ingrid Levy
- KidZ Health Castle, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel , Brussels, Belgium
| | - Mariam Ouald Chaib
- KidZ Health Castle, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel , Brussels, Belgium
| | - Yvan Vandenplas
- KidZ Health Castle, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel , Brussels, Belgium
| |
Collapse
|
15
|
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: 23.6] [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
|
16
|
Castejón-Castejón M, Murcia-González M, Martínez Gil J, Todri J, Suárez Rancel M, Lena O, Chillón-Martínez R. Effectiveness of craniosacral therapy in the treatment of infantile colic. A randomized controlled trial. Complement Ther Med 2019; 47:102164. [DOI: 10.1016/j.ctim.2019.07.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 07/25/2019] [Accepted: 07/29/2019] [Indexed: 01/20/2023] Open
|
17
|
Lazou Ahrén I, Berggren A, Teixeira C, Martinsson Niskanen T, Larsson N. Evaluation of the efficacy of Lactobacillus plantarum HEAL9 and Lactobacillus paracasei 8700:2 on aspects of common cold infections in children attending day care: a randomised, double-blind, placebo-controlled clinical study. Eur J Nutr 2019; 59:409-417. [PMID: 31734734 PMCID: PMC7000506 DOI: 10.1007/s00394-019-02137-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 11/04/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND The combination of Lactobacillus plantarum HEAL9 and Lactobacillus paracasei 8700:2 (commercially available as Probi Defendum®) has previously been reported to reduce the incidence, duration and severity of naturally acquired common colds in adults. The aim of the present study was to evaluate the impact of Probi Defendum® on aspects of common cold in healthy children 1-6 years of age attending day care. METHODS A total of 131 children, out of the planned 320, were recruited into the study during 1 common cold season and randomised to consume once daily either 109 CFU (colony forming units) of the probiotic product or placebo. Due to unforeseen reasons, the recruitment of more children did not continue beyond the first cold season. RESULTS There were 106 children that completed the study out of the 131 randomised. Daily consumption of the probiotic product for a period of 3 months significantly reduced the severity of the symptom "nasal congestion/runny nose" with a mean severity score for the whole study period of 7.5 ± 9.7 in the probiotic group and 13.9 ± 15.2 in the placebo (p < 0.05). Moreover, significantly less concomitant medication was used in the probiotic group. When the data were projected to a larger population corresponding to the originally estimated sample size, the results were in favour of the probiotic group regarding the reduced absence from day care (p < 0.05), reduced mean total severity per day in the reported episodes (p < 0.05) and reduced severity of the symptom "crying more than usual" (p < 0.05). CONCLUSION Intake of Probi Defendum® once daily for a period of 3 months was beneficial to children and reduced the severity of common colds.
Collapse
|
18
|
Endothelial Function is improved by Inducing Microbial Polyamine Production in the Gut: A Randomized Placebo-Controlled Trial. Nutrients 2019; 11:nu11051188. [PMID: 31137855 PMCID: PMC6566626 DOI: 10.3390/nu11051188] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/23/2019] [Accepted: 05/23/2019] [Indexed: 12/20/2022] Open
Abstract
Recently, it was demonstrated that spermidine-induced autophagy reduces the risk of cardiovascular disease in mice. Intestinal bacteria are a major source of polyamines, including spermidine. We previously reported that the intake of both Bifidobacterium animalis subsp. lactis (Bifal) and arginine (Arg) increases the production of putrescine, a spermidine precursor, in the gut. Here, we investigated the effects of Bifal and Arg consumption on endothelial function in healthy subjects. Healthy individuals with body mass index (BMI) near the maximum value in the “healthy” range (BMI: 25) (n = 44) were provided normal yogurt containing Bifal and Arg (Bifal + Arg YG) or placebo (normal yogurt) for 12 weeks in this randomized, double-blinded, placebo-controlled, parallel-group comparative study. The reactive hyperemia index (RHI), the primary outcome, was measured using endo-peripheral arterial tone (EndoPAT). The change in RHI from week 0 to 12 in the Bifal + Arg YG group was significantly higher than that in the placebo group, indicating that Bifal + Arg YG intake improved endothelial function. At week 12, the concentrations of fecal putrescine and serum putrescine and spermidine in the Bifal + Arg YG group were significantly higher than those in the placebo group. This study suggests that consuming Bifal + Arg YG prevents or reduces the risk of atherosclerosis.
Collapse
|
19
|
Sanders A, Rackers H, Kimmel M. A role for the microbiome in mother-infant interaction and perinatal depression. Int Rev Psychiatry 2019; 31:280-294. [PMID: 30784334 DOI: 10.1080/09540261.2018.1548431] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Perinatal depression is a significant public health problem, due to its negative impact on maternal well-being and long-term adverse effects for children. Mother-infant interaction and maternal responsiveness and sensitivity are a hypothesized mechanism by which perinatal depression effects child development, and increasing research in the microbiota-gut-brain axis may provide a new avenue of investigation. There is limited efficacy for treatment of perinatal depression for improving the mother-infant relationship and child outcomes. The maternal microbiota may be the basis of child outcomes through foetal programming and sharing of microbes between mother and infant. There is evidence that less diversity of the intestinal microbial community is associated with neuropsychiatric disorders, including depression and anxiety in mothers and offspring. Assessing the maternal and child's microbial communities may be an important missing component in mother-infant attachment-based therapies during treatment of perinatal depression. Probiotics and prebiotics require further research as additions to mother-infant interventions. Further research may enable identification of bacterial genes that indicate specific pathways that could be targeted to improve outcomes for mother and child.
Collapse
Affiliation(s)
- Amanda Sanders
- a Department of Psychiatry , University of North Carolina at Chapel Hill , Chapel Hill , NC , USA
| | - Hannah Rackers
- a Department of Psychiatry , University of North Carolina at Chapel Hill , Chapel Hill , NC , USA
| | - Mary Kimmel
- a Department of Psychiatry , University of North Carolina at Chapel Hill , Chapel Hill , NC , USA
| |
Collapse
|
20
|
The Effect of Synbiotic Supplementation on Growth Parameters in Mild to Moderate FTT Children Aged 2–5 Years. Probiotics Antimicrob Proteins 2019; 12:119-124. [DOI: 10.1007/s12602-018-9508-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
21
|
Seo MK, Park EJ, Ko SY, Choi EW, Kim S. Therapeutic effects of kefir grain Lactobacillus-derived extracellular vesicles in mice with 2,4,6-trinitrobenzene sulfonic acid-induced inflammatory bowel disease. J Dairy Sci 2018; 101:8662-8671. [PMID: 30100498 DOI: 10.3168/jds.2018-15014] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 06/17/2018] [Indexed: 01/19/2023]
Abstract
Kefir is a fermented product from yeast and lactic acid bacteria, and has been associated with various health benefits including relieving inflammatory bowel disease. Recently, it has been shown that gram-positive bacteria produce extracellular vesicles (EV). The EV could be appearing as potentially important mediators of cell to cell interaction. In this study, we explored the role of kefir grain Lactobacillus-derived EV in modulating inflammation responses via alleviating the production of inflammatory cytokines in tumor necrosis factor-α (TNF-α)-induced inflammation in Caco-2 cells and the 2,4,6-trinitrobenzene sulfonic acid-induced inflammatory bowel disease mouse model. Kefir-derived Lactobacillus EV were isolated by ultracentrifugation of the culture medium of 3 different kefir-derived strains (i.e., Lactobacillus kefir, Lactobacillus kefiranofaciens, and Lactobacillus kefirgranum). Nanoparticle tracking analysis showed that the size of isolated kefir-derived Lactobacillus EV was within 80 to 400 nm, and kefir-derived Lactobacillus EV uptake into recipient Caco-2 cells was confirmed by fluorescence labeling. Treatment of each kefir-derived Lactobacillus EV onto TNF-α-stimulated Caco-2 cells significantly reduced the level of both mRNA expression and secretion of IL-8, and Western blot analysis revealed that such an effect was related to inhibition of TNF-α signaling mediated by reducing the phosphorylation of p65, a subunit of NF-kB. Subsequent administration of kefir-derived Lactobacillus EV into inflammatory bowel disease-induced mice significantly alleviated the body weight loss and rectal bleeding, and enhanced stool consistency. Histological examination showed that kefir-derived Lactobacillus EV substantially reduced the infiltration of transmural leukocytes and loss of goblet cells within the colon, and the serum level of myeloperoxidase was significantly lower in the EV-treated group than control group. Our study demonstrates that kefir-derived Lactobacillus EV can be potentially used for developing innovative strategies for alleviating inflammatory bowel disease.
Collapse
Affiliation(s)
- M K Seo
- Prostemics Research Institute, Seongdong-gu, Seoul 04778, South Korea
| | - E J Park
- Prostemics Research Institute, Seongdong-gu, Seoul 04778, South Korea
| | - S Y Ko
- Prostemics Research Institute, Seongdong-gu, Seoul 04778, South Korea
| | - E W Choi
- Prostemics Research Institute, Seongdong-gu, Seoul 04778, South Korea
| | - S Kim
- Prostemics Research Institute, Seongdong-gu, Seoul 04778, South Korea.
| |
Collapse
|
22
|
Solano-Aguilar G, Shea-Donohue T, Madden KB, Quinoñes A, Beshah E, Lakshman S, Xie Y, Dawson H, Urban JF. Bifidobacterium animalis subspecies lactis modulates the local immune response and glucose uptake in the small intestine of juvenile pigs infected with the parasitic nematode Ascaris suum. Gut Microbes 2018; 9:422-436. [PMID: 30024817 PMCID: PMC6219643 DOI: 10.1080/19490976.2018.1460014] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
An evaluation of a localized intestinal allergic type-2 response concomitant with consumption of probiotic bacteria is not well documented. This study investigated the effect of feeding probiotic Bifidobacterium animalis subspecies lactis (Bb12) or a placebo in weaned pigs that were also inoculated with Ascaris suum (A. suum) eggs to induce a strong Th2-dependent allergic type 2 immune response. Sections of jejunal mucosa were mounted in Ussing chambers to determine changes in permeability and glucose absorption, intestine and liver samples were collected for analysis of type-2 related gene expression, jejunum examined histologically, and sera and intestinal fluid were assayed for parasite antigen specific antibody. The prototypical parasite-induced secretory response to histamine and reduced absorption of glucose in the jejunum were attenuated by feeding Bb12 without a change in mucosal resistance. Parasite antigen-specific IgA response in the serum and IgG1 and IgG2 response in the ileal fluid were significantly increased in A. suum-infected pigs treated with Bb12 compared to infected pigs given the placebo. Ascaris suum-induced eosinophilia in the small intestinal mucosa was inhibited by Bb12 treatment without affecting the normal expulsion of A. suum 4th stage larvae (L4) or the morphometry of the intestine. Expression of genes associated with Th1/Th2 cells, Treg cells, mast cells, and physiological function in the intestine were modulated in A. suum infected-pigs treated with Bb12. These results suggested that Bb12 can alter local immune responses and improve intestinal function during a nematode infection by reducing components of a strong allergenic type-2 response in the pig without compromising normal parasite expulsion.
Collapse
Affiliation(s)
- Gloria Solano-Aguilar
- United States Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet Genomics and Immunology Laboratory, Beltsville, MD,CONTACT Gloria Solano-Aguilar 10300 Baltimore Avenue, BARC-East. Bldg 307C, Room 225, Beltsville, MD 20705, USA
| | - Terez Shea-Donohue
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD
| | - Kathleen B. Madden
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, MD
| | | | - Ethiopia Beshah
- United States Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet Genomics and Immunology Laboratory, Beltsville, MD
| | - Sukla Lakshman
- United States Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet Genomics and Immunology Laboratory, Beltsville, MD
| | - Yue Xie
- Department of Parasitology, Sichuan Agricultural University, College of Veterinary Medicine, Sichuan, China
| | - Harry Dawson
- United States Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet Genomics and Immunology Laboratory, Beltsville, MD
| | - Joseph F. Urban
- United States Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet Genomics and Immunology Laboratory, Beltsville, MD
| |
Collapse
|
23
|
Laursen RP, Hojsak I. Probiotics for respiratory tract infections in children attending day care centers-a systematic review. Eur J Pediatr 2018; 177:979-994. [PMID: 29752587 DOI: 10.1007/s00431-018-3167-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 04/30/2018] [Accepted: 05/02/2018] [Indexed: 12/15/2022]
Abstract
Probiotics have been suggested to have a preventive effect on respiratory tract infections (RTIs), but limited evidence exist on strain-specific effects. The main aim of this systematic review and meta-analysis was to evaluate strain-specific probiotic effects on RTIs in children attending day care. We included 15 RCTs with 5121 children in day care settings (aged 3 months to 7 years), but due to high diversity in reported outcomes, different number of RCTs were available for evaluated outcomes. Twelve RCTs (n = 4527) reported results which could be compared in at least one outcome of the meta-analysis. Compared to placebo, Lactobacillus rhamnosus GG (LGG) significantly reduced duration of RTIs (three RCTs, n = 1295, mean difference - 0.78 days, 95% confidence interval (CI) - 1.46; - 0.09), whereas no effect was found on other evaluated outcomes. Based on the results from two studies (n = 343), Bifidobacterium animalis subsp. lactis BB-12 showed no effect on duration of RTIs or on absence from day care. Meta-analyses on other strains or their combination were not possible due to limited data and different outcome measures.Conclusion: LGG is modestly effective in decreasing the duration of RTIs. More RCTs investigating specific probiotic strains or their combinations in prevention of RTIs are needed. What is known: • Previously published systematic reviews have suggested that probiotics may have a preventive effect on respiratory infections, but limited data exist on strain specific effects. What is new: • This systematic review showed that use of Lactobacillus rhamnosus GG modestly reduces the duration of respiratory tract infections.
Collapse
Affiliation(s)
- Rikke Pilmann Laursen
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Iva Hojsak
- Children's Hospital Zagreb, School of Medicine, University of Zagreb, Zagreb, Croatia. .,School of Medicine, University J.J. Strossmayer, Osijek, Croatia. .,Referral Center for Pediatric Gastroenterology and Nutrition, Children's Hospital Zagreb, Klaićeva 16, 10000, Zagreb, Croatia.
| |
Collapse
|
24
|
Bifidobacterium longum subsp infantis CECT7210-supplemented formula reduces diarrhea in healthy infants: a randomized controlled trial. Pediatr Res 2018. [PMID: 29538368 DOI: 10.1038/pr.2018.34] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BackgroundIntestinal microbiota of breast-fed infants is plenty of beneficial bifidobacteria. We aimed to determine whether an infant formula supplemented with probiotic Bifidobacterium longum subsp. infantis CECT7210 (B. infantis IM1) is effective at reducing diarrhea incidence in healthy term infants.MethodsDouble-blinded, randomized, multicenter, controlled clinical trial, where formula-fed infants (<3 months) received an infant formula supplemented (Probiotic) or not (Control) with 107 cfu/g of B. infantis IM1 over 12 weeks. Diarrheas, growth, digestive symptoms, stool bifidobacteria, and microbiota were assessed.ResultsIn all, 97 (Control) and 93 (Probiotic) infants were randomized, and 78 (Control) and 73 (Probiotic) completed the 12 week-follow-up. In the overall study period, a median of 0.29±1.07 and 0.05±0.28 diarrhea events/infant was observed in the Control and Probiotic groups, respectively (P=0.059). This trend to less diarrhea episodes in the Probiotic group reached statistical significance at 8 weeks (0.12±0.47 vs. 0.0±0.0 events/infant, P=0.047). Constipation incidence was higher (odds ratio (OR) 2.67 (1.09-6.50)) and stool frequency lower (2.0±1.0 vs. 2.6±1.3 stools/day, P=0.038) in the Control group after 4 weeks. No differences were found at other time points nor in other digestive symptoms, growth, or formula intake.ConclusionA B. infantis IM1-supplemented infant formula may reduce diarrhea episodes, being safe, well tolerated, and associated with lower constipation prevalence.
Collapse
|
25
|
Zoumpopoulou G, Tsakalidou E, Thomas L. An Overview of Probiotic Research. PROBIOTIC DAIRY PRODUCTS 2017:293-357. [DOI: 10.1002/9781119214137.ch8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
26
|
Watkins C, Stanton C, Ryan CA, Ross RP. Microbial Therapeutics Designed for Infant Health. Front Nutr 2017; 4:48. [PMID: 29124056 PMCID: PMC5662644 DOI: 10.3389/fnut.2017.00048] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 09/26/2017] [Indexed: 12/13/2022] Open
Abstract
Acknowledgment of the gut microbiome as a vital asset to health has led to multiple studies attempting to elucidate its mechanisms of action. During the first year of life, many factors can cause fluctuation in the developing gut microbiome. Host genetics, maternal health status, mode of delivery, gestational age, feeding regime, and perinatal antibiotic usage, are known factors which can influence the development of the infant gut microbiome. Thus, the microbiome of vaginally born, exclusively breastfed infants at term, with no previous exposure to antibiotics, either directly or indirectly from the mother, is to be considered the "gold standard." Moreover, the use of prebiotics as an aid for the development of a healthy gut microbiome is equally as important in maintaining gut homeostasis. Breastmilk, a natural prebiotic source, provides optimal active ingredients for the growth of beneficial microbial species. However, early life disorders such as necrotising enterocolitis, childhood obesity, and even autism have been associated with an altered/disturbed gut microbiome. Subsequently, microbial therapies have been introduced, in addition to suitable prebiotic ingredients, which when administered, may aid in the prevention of a microbial disturbance in the gastrointestinal tract. The aim of this mini-review is to highlight the beneficial effects of different probiotic and prebiotic treatments in early life, with particular emphasis on the different conditions which negatively impact microbial colonisation at birth.
Collapse
Affiliation(s)
- Claire Watkins
- APC Microbiome Institute, University College Cork, Cork, Ireland
- Teagasc Food Research Centre, Fermoy, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Institute, University College Cork, Cork, Ireland
- Teagasc Food Research Centre, Fermoy, Ireland
| | - C. Anthony Ryan
- APC Microbiome Institute, University College Cork, Cork, Ireland
- Department of Neonatology, Cork University Maternity Hospital, Cork, Ireland
| | - R. Paul Ross
- APC Microbiome Institute, University College Cork, Cork, Ireland
- Teagasc Food Research Centre, Fermoy, Ireland
- School of Science, Engineering and Food Science, University College Cork, Cork, Ireland
| |
Collapse
|
27
|
Skórka A, Pieścik-Lech M, Kołodziej M, Szajewska H. To add or not to add probiotics to infant formulae? An updated systematic review. Benef Microbes 2017; 8:717-725. [DOI: 10.3920/bm2016.0233] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We updated evidence on the effects of the administration of probiotic-supplemented infant formulae (IF) compared with unsupplemented IF. Five databases were searched up to September 2016 for randomised controlled trials. Twenty publications were identified, including five new RCTs. Supplementation of IF with Bifidobacterium lactis Bb12, either alone or with Streptococcus thermophilus, had no effect on growth, respiratory illness, antibiotic use, stool frequency or consistency. However, there was a significant reduction in the number of episodes of gastrointestinal infections (Bb12) and a lower frequency of colic or irritability (when both strains were used). Lactobacillus johnsonii La1 had no effect on growth, gastrointestinal infections, or respiratory illness episodes. There were no effects of supplementation of IF with Bifidobacterium longum BL999, alone or with Lactobacillus rhamnosus LPR. L. rhamnosus GG was associated with better growth; it had no effect on colic/crying, or irritability, and it was associated with greater indexes of loose stools and a higher defecation frequency. Lactobacillus reuteri ATCC 55730 had no effect on growth, colic, crying, irritability, respiratory illness, antibiotic use, stool frequency, or stool consistency; however, it reduced the number of episodes of diarrhoea. L. reuteri DSM 17938 had no effect on growth, night-time sleeping, or flatulence, but it reduced the number of spitting episodes. Lactobacillus salivarius CEC5713 had no effect on growth, colic, crying, or irritability; however, it resulted in a significant reduction in the rate of diarrhoea and the number of episodes of respiratory symptoms. In conclusion, the administration of probiotic-supplemented formulae to healthy infants does not raise safety concerns with regard to growth and adverse effects. Some beneficial clinical effects are possible; however, there is no existing robust evidence to recommend their routine use. The latter conclusion may reflect the small amount of data on a specific probiotic strain(s) and outcomes, rather than a genuine lack of an effect.
Collapse
Affiliation(s)
- A. Skórka
- Department of Paediatrics, Medical University of Warsaw, Żwirki iWigury 63A, 02-091 Warsaw, Poland
| | - M. Pieścik-Lech
- Department of Paediatrics, Medical University of Warsaw, Żwirki iWigury 63A, 02-091 Warsaw, Poland
| | - M. Kołodziej
- Department of Paediatrics, Medical University of Warsaw, Żwirki iWigury 63A, 02-091 Warsaw, Poland
| | - H. Szajewska
- Department of Paediatrics, Medical University of Warsaw, Żwirki iWigury 63A, 02-091 Warsaw, Poland
| |
Collapse
|
28
|
Streptococcus thermophilus: From yogurt starter to a new promising probiotic candidate? J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.07.038] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
29
|
Ley D, Desseyn JL, Mischke M, Knol J, Turck D, Gottrand F. Early-life origin of intestinal inflammatory disorders. Nutr Rev 2017; 75:175-187. [PMID: 28340001 DOI: 10.1093/nutrit/nuw061] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
A growing body of evidence supports the concept of perinatal programming through which the perinatal environment affects the development of the fetus and infant, thereby modifying the risk profile for disease later in life. Increasing attention is focusing on the role of the early environment in the development of chronic intestinal disorders. Epidemiological studies have highlighted the link between perinatal factors, such as breastfeeding, cesarean delivery, and antibiotic use, and an increased risk for inflammatory bowel disease and/or celiac disease. These links are consistent with the concept of perinatal programming of intestinal inflammatory disorders. Animal models have shown that the early-life environment affects the development of the gastrointestinal tract, but further experimental studies are needed to confirm the long-term effects of the perinatal environment on susceptibility to chronic intestinal disorders later in life. Changes in the development and composition of the intestinal microbiota as well as epigenetic changes are emerging as key mechanisms through which the perinatal environment determines susceptibility to intestinal inflammatory disorders.
Collapse
Affiliation(s)
- Delphine Ley
- Lille Inflammation Research International Center (LIRIC) - UMR 995 Inserm, University Lille, CHU Lille, Lille, France
| | - Jean-Luc Desseyn
- Lille Inflammation Research International Center (LIRIC) - UMR 995 Inserm, University Lille, CHU Lille, Lille, France
| | | | - Jan Knol
- Nutricia Research, Utrecht, The Netherlands.,Laboratory of Microbiology, Wageningen University, The Netherlands
| | - Dominique Turck
- Lille Inflammation Research International Center (LIRIC) - UMR 995 Inserm, University Lille, CHU Lille, Lille, France
| | - Frédéric Gottrand
- Lille Inflammation Research International Center (LIRIC) - UMR 995 Inserm, University Lille, CHU Lille, Lille, France
| |
Collapse
|
30
|
Hojsak I. Probiotics in Children: What Is the Evidence? Pediatr Gastroenterol Hepatol Nutr 2017; 20:139-146. [PMID: 29026729 PMCID: PMC5636929 DOI: 10.5223/pghn.2017.20.3.139] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 07/31/2017] [Indexed: 01/05/2023] Open
Abstract
The number of papers discussing probiotics increases tremendously that limits the possibility for primary care physicians and clinicians to stay updated. Therefore, the aim of this paper will be to summarize available evidence of probiotic use in well-defined clinical indications of importance for pediatricians. Based on currently available evidence certain probiotic strains (Lactobacillus rhamnosus GG [LGG] and Saccharomyces boulardii) have proven effect in the treatment of acute gastroenteritis and prevention of antibiotic associated diarrhea. Furthermore, LGG was proven to be effective in prevention of nosocomial diarrhea and respiratory tract infection in day care centers. In conclusion, not all probiotic strains have same efficacy for all clinical indications, therefore, only strains with proven efficacy and safety should be recommended.
Collapse
Affiliation(s)
- Iva Hojsak
- Children's Hospital Zagreb, Zagreb, Croatia.,University of Zagreb, School of Medicine, Zagreb, Croatia.,University Josip Juraj Strossmayer, School of Medicine Osijek, Osijek, Croatia
| |
Collapse
|
31
|
Manzano S, De Andrés J, Castro I, Rodríguez J, Jiménez E, Espinosa-Martos I. Safety and tolerance of three probiotic strains in healthy infants: a multi-centre randomized, double-blind, placebo-controlled trial. Benef Microbes 2017; 8:569-578. [DOI: 10.3920/bm2017.0009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Some strains of species belonging to the genera Bifidobacterium and Lactobacillus are used in order to maintain health. Although these organisms have a long record of safe use, it is important to assess their safety and tolerance in potentially vulnerable populations, such as infants. The objective of this study was to evaluate the safety and tolerance of three probiotic strains (Bifidobacterium longum subsp. infantis R0033, Bifidobacterium bifidum R0071 and Lactobacillus helveticus R0052) in healthy infants aged 3 to 12 months. A multi-centre randomized, double-blind, placebo-controlled intervention study with 221 healthy full-term infants was conducted. Infants received either a placebo or one of the 3 probiotic strains (3×109 cfu) daily during an 8 week intervention period. Growth (weight, height and head circumference), adverse events (AEs)/serious adverse events (SAEs), concentrations of D-lactic acid in urine samples, characteristics of the stools and use of medication were collected for safety evaluation. All 4 groups were homogeneous with respect to age, gender, feeding type, ethnicity, height, weight and head circumference at the start of the study. The results showed that changes in growth (weight, height and head circumference) were equivalent in all 4 groups. No SAEs were reported. Total number of AEs recorded was equivalent in all groups. Thus, the use of B. infantis R0033, L. helveticus R0052 and B. bifidum R0071 in infancy is safe, and well tolerated.
Collapse
Affiliation(s)
- S. Manzano
- Dpto. Nutrición, Bromatología y Tecnología de los Alimentos, Universidad Complutense de Madrid, Av. Puerta de hierro s/n, 28040 Madrid, Spain
- Probisearch S.L.U., C/ Santiago Grisolía, 2, 28760 Tres Cantos, Spain
| | - J. De Andrés
- Dpto. Nutrición, Bromatología y Tecnología de los Alimentos, Universidad Complutense de Madrid, Av. Puerta de hierro s/n, 28040 Madrid, Spain
| | - I. Castro
- Dpto. Nutrición, Bromatología y Tecnología de los Alimentos, Universidad Complutense de Madrid, Av. Puerta de hierro s/n, 28040 Madrid, Spain
| | - J.M. Rodríguez
- Dpto. Nutrición, Bromatología y Tecnología de los Alimentos, Universidad Complutense de Madrid, Av. Puerta de hierro s/n, 28040 Madrid, Spain
- Probisearch S.L.U., C/ Santiago Grisolía, 2, 28760 Tres Cantos, Spain
| | - E. Jiménez
- Dpto. Nutrición, Bromatología y Tecnología de los Alimentos, Universidad Complutense de Madrid, Av. Puerta de hierro s/n, 28040 Madrid, Spain
- Probisearch S.L.U., C/ Santiago Grisolía, 2, 28760 Tres Cantos, Spain
| | - I. Espinosa-Martos
- Dpto. Nutrición, Bromatología y Tecnología de los Alimentos, Universidad Complutense de Madrid, Av. Puerta de hierro s/n, 28040 Madrid, Spain
- Probisearch S.L.U., C/ Santiago Grisolía, 2, 28760 Tres Cantos, Spain
| |
Collapse
|
32
|
Xinias I, Analitis A, Mavroudi A, Roilides I, Lykogeorgou M, Delivoria V, Milingos V, Mylonopoulou M, Vandenplas Y. Innovative Dietary Intervention Answers to Baby Colic. Pediatr Gastroenterol Hepatol Nutr 2017; 20:100-106. [PMID: 28730134 PMCID: PMC5517376 DOI: 10.5223/pghn.2017.20.2.100] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 02/04/2017] [Accepted: 02/14/2017] [Indexed: 01/20/2023] Open
Abstract
PURPOSE The purpose of this paper is to evaluate the efficacy of a lactose- reduced synbiotic partial whey hydrolysate in formula fed infants presenting with colic and the impact of this dietary intervention in mean crying time and quality of life. METHODS Forty infants with infantile colic were treated during one month with parental reassurance and the intervention formula (partial whey hydrolysate, reduced lactose, Bifidobacterium lactis BB12 and galacto-oligosaccharides) and were compared to a control group of 20 infants with infantile colic treated with parental reassurance and a standard infant formula. Parents completed a quality of life (QoL) questionnaire assessing the burden of infantile colic. Wilcoxon test, t-test and Mann-Whitney test were used to compare QoL scores before and after intervention as well as between the intervention and control group. RESULTS At inclusion, duration of crying did not differ between both groups. Crying duration decreased with 2.7 hours (from 3.2 to 0.5 hours) in the intervention group while duration of crying decreased only with 1.2 hours in the control group (p<0.001). Stool composition became looser in the intervention group, but defecation frequency did not change. The median scores of the QoL questionnaire improved significantly in the intervention group for all parameters. In the control group, parameters improved significantly also but not for the parent-child and social interaction. The score changes were significantly greater in the intervention than in the control group. CONCLUSION The intervention formula (partial whey hydrolysate, synbiotic, reduced lactose) significantly reduced the duration of crying and improved QoL of the parents and infants.
Collapse
Affiliation(s)
- Ioannis Xinias
- 3rd Pediatric Department, Hippocration Hospital, Thessaloniki, Greece
| | - A Analitis
- Department of Hygiene, Epidemiology and Medical Statistics, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Antigoni Mavroudi
- 3rd Pediatric Department, Hippocration Hospital, Thessaloniki, Greece
| | - Ioannis Roilides
- 3rd Pediatric Department, Hippocration Hospital, Thessaloniki, Greece
| | - Maria Lykogeorgou
- 3rd Pediatric Department, Hippocration Hospital, Thessaloniki, Greece
| | - Varvara Delivoria
- 3rd Pediatric Department, Hippocration Hospital, Thessaloniki, Greece
| | - Vasilis Milingos
- 3rd Pediatric Department, Hippocration Hospital, Thessaloniki, Greece
| | | | - Yvan Vandenplas
- Department of Pediatrics, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
33
|
Xiao L, Ding G, Ding Y, Deng C, Ze X, Chen L, Zhang Y, Song L, Yan H, Liu F, Ben X. Effect of probiotics on digestibility and immunity in infants: A study protocol for a randomized controlled trial. Medicine (Baltimore) 2017; 96:e5953. [PMID: 28383398 PMCID: PMC5411182 DOI: 10.1097/md.0000000000005953] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 01/03/2017] [Indexed: 12/28/2022] Open
Abstract
The gastrointestinal (GI) tract of a fetus in utero is sterile but it becomes colonized with environmental microorganisms shortly after birth. Since the gut microbiota undergoes substantial changes in early life, healthy gut microflora is essential to an infant's gut health and immune system and probably also has an effect on overall health status in later life. Probiotics, defined as viable microbial preparations that have a beneficial effect on the health of the host, represent a rapidly expanding field. Although randomized controlled trials using probiotics in infants have shown promising results in the prevention and treatment of common diseases such as diarrhea and allergy, little is known about whether probiotics could offer benefits to healthy infants. We have designed a randomized controlled trial to test the hypothesis that an oral preparation of probiotics is superior to placebo in improving digestive and immune function in healthy infants.The trial will be a randomized, double-blind, placebo-controlled, 2-parallel-group study in Shanghai, China. After a 2-week run-in period, 200 exclusively formula-fed healthy infants aged 4 to 6 months will be randomly allocated to receive either a probiotic product containing Bifidobacterium infantis R0033, Bifidobacterium bifidum R0071, and Lactobacillus helveticus R0052 or an identical placebo once daily for 4 weeks and will be followed up for 8 weeks. The duration of the subject's participation will be 14 weeks, with a total of 5 visits: inclusion (Visit 1, Day 1), start of intervention (V2, D15), end of intervention (V3, D44), and follow-up (V4 and V5, D72 and D100). Stool and saliva samples will be collected at the first 3 visits to measure microbial populations and secretory immunoglobulin A (SIgA), respectively. Physical examination will be performed at each visit, and tolerance records will be completed 1 day prior to each visit. The primary endpoints will be the changes in the composition of fecal microbiota, particularly the Bifidobacterium bifidum population. The secondary endpoints will include the change in salivary SIgA level, growth parameters, digestive tolerance, and adverse events.An effective, practical, and acceptable probiotic intervention in manipulating the gut microbiota and boosting the immune system in formula-fed infants would represent a major clinical advance. The administration of probiotic supplementation or follow-on formula to infant may be associated with some clinic benefits.
Collapse
Affiliation(s)
- Lingli Xiao
- Department of Neonatology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guodong Ding
- Department of Pediatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yifang Ding
- Department of Neonatology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chaoming Deng
- Biostime Institute for Nutrition and Care, Biostime Co. Ltd., Guangzhou, China
| | - Xiaolei Ze
- Biostime Institute for Nutrition and Care, Biostime Co. Ltd., Guangzhou, China
| | - Liang Chen
- Biostime Institute for Nutrition and Care, Biostime Co. Ltd., Guangzhou, China
| | - Yao Zhang
- Department of Food Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Lihua Song
- Department of Food Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Hongli Yan
- Department of Laboratory Medicine, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Fang Liu
- Department of Pediatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoming Ben
- Department of Neonatology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
34
|
Barnes D, Yeh AM. Bugs and Guts: Practical Applications of Probiotics for Gastrointestinal Disorders in Children. Nutr Clin Pract 2016; 30:747-59. [PMID: 26538058 DOI: 10.1177/0884533615610081] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Probiotics are foods or products that contain live microorganisms that benefit the host when administered. In this clinical review, we evaluate the literature associated with using probiotics in common pediatric gastrointestinal disorders, focusing specifically on antibiotic-associated diarrhea, acute gastroenteritis, Clostridium difficile infection (CDI), colic, inflammatory bowel disease, and functional gastrointestinal diseases. Meta-analysis of several randomized controlled trials have confirmed benefit for the administration of Lactobacillus rhamnosus GG and Saccharomyces boulardii to prevent antibiotic-associated diarrhea and to treat acute infectious diarrhea. Individual studies have also suggested benefit of probiotics to prevent acute gastroenteritis and serve as an adjunct in ulcerative colitis, pouchitis, antibiotic-associated diarrhea, CDI, functional abdominal pain, irritable bowel syndrome, and colic in breastfed babies. Although promising, larger well-designed studies need to confirm these findings. There is currently insufficient evidence to recommend probiotics for the treatment of constipation-predominant irritable bowel syndrome or Crohn's disease.
Collapse
Affiliation(s)
- Danielle Barnes
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Stanford Children's Health, Stanford University, Palo Alto, California
| | - Ann Ming Yeh
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Stanford Children's Health, Stanford University, Palo Alto, California
| |
Collapse
|
35
|
Fields D, Czerkies L, Sun S, Storm H, Saavedra J, Sorensen R. A Randomized Controlled Trial Assessing Growth of Infants Fed a 100% Whey Extensively Hydrolyzed Formula Compared With a Casein-Based Extensively Hydrolyzed Formula. Glob Pediatr Health 2016; 3:2333794X16636613. [PMID: 27336009 PMCID: PMC4905054 DOI: 10.1177/2333794x16636613] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 01/20/2016] [Accepted: 01/30/2016] [Indexed: 11/30/2022] Open
Abstract
This study compared the growth of healthy infants fed a hypoallergenic 100% whey-based extensively hydrolyzed formula (EHF) with Bifidobacterium lactis (test) with that of infants fed an extensively hydrolyzed casein formula (control). Formula-fed infants (14 ± 3 days) were randomized to test or control groups until 112 days of age. Anthropometrics were assessed at 14, 28, 56, 84, and 112 days, and daily records were kept for 2 days prior to study visits. Serum albumin and plasma amino acids at 84 days were assessed in a subset. A total of 282 infants were randomized (124 test, 158 control). Significantly more infants dropped out of the control (56%) as compared with the test (41%) group. Mean daily weight gain was significantly higher in the test group compared with the control group (27.95 ± 5.91 vs 25.93 ± 6.12 g/d; P = .027) with the test group reporting significantly fewer stools (2.2 vs 3.6 stools/d; P < .0001). The control group reported significantly more days with >3 loose stools/d and a higher incidence of vomiting as compared with the test group. There were no differences in gas, mood, sleep, or serum albumin. Plasma arginine and valine were significantly lower in the test group, whereas leucine and lysine were higher; all values were within normal limits. Significantly more adverse events attributed to the study formula were reported in the control group. The 100% whey-based hypoallergenic EHF containing Bifidobacterium lactis and medium chain triglycerides supported growth of healthy infants. Future studies on the application of this formula in clinically indicated populations are warranted.
Collapse
Affiliation(s)
- David Fields
- University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | - Shumei Sun
- Virginia Commonwealth University, Richmond, VA, USA
| | | | - José Saavedra
- Nestlé Nutrition, Vevey, Switzerland
- José M. Saavedra, Nestlé Nutrition, Av Nestlé 55, Vevey, 1800, Switzerland.
| | | |
Collapse
|
36
|
Chen YL, Liao FH, Lin SH, Chien YW. A Prebiotic Formula Improves the Gastrointestinal Bacterial Flora in Toddlers. Gastroenterol Res Pract 2016; 2016:3504282. [PMID: 27403155 PMCID: PMC4923535 DOI: 10.1155/2016/3504282] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 04/16/2016] [Accepted: 05/16/2016] [Indexed: 12/26/2022] Open
Abstract
We aimed to investigate the effect of enriched 3-prebiotic formula (including inulin, fructooligosaccharides, and galactooligosaccharides) on toddler gut health by measuring fecal microbiota. Our results revealed that the consumption of 3-prebiotic formula three times per day giving total intake of 1.8 g prebiotic ingredients significantly showed the increased number of probiotic Bifidobacterium spp. colonies and the reduced populations of both C. perfringens and total anaerobic bacteria on the fecal bacterial flora in toddlers at 18~36 months. In addition, total organic acids in the fecal samples significantly increased which improves the utilization of bifidus under acidic conditions after consumption of the 3-prebiotic formula. Therefore, using the formula enriched with prebiotic may maintain gut health in toddlers.
Collapse
Affiliation(s)
- Ya-Ling Chen
- Department of Nutrition and Health Sciences, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan
| | - Fang-Hsuean Liao
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 11014, Taiwan
| | - Shyh-Hsiang Lin
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 11014, Taiwan
| | - Yi-Wen Chien
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 11014, Taiwan
| |
Collapse
|
37
|
Bifidobacterium animalis subsp. lactis in prevention of common infections in healthy children attending day care centers – Randomized, double blind, placebo-controlled study. Clin Nutr 2016; 35:587-91. [DOI: 10.1016/j.clnu.2015.05.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 03/01/2015] [Accepted: 05/12/2015] [Indexed: 11/17/2022]
|
38
|
Weizman Z. The role of probiotics and prebiotics in the prevention of infections in child day-care centres. Benef Microbes 2016; 6:181-3. [PMID: 25391348 DOI: 10.3920/bm2014.0101] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Infants and children attending day-care centres demonstrate a notably higher risk of gastrointestinal as well as of respiratory tract infections. The present short review evaluates various well-controlled clinical trials analysing the effect of probiotics and prebiotics in the prevention of such infections. In most of the 12 studies identified, probiotic supplementation was found to be a safe and effective therapeutic tool in preventing gastrointestinal and respiratory infection in this population.
Collapse
Affiliation(s)
- Z Weizman
- Pediatric Gastroenterology and Nutrition Unit, Soroka University Medical Center, Ben-Gurion University, P.O. Box 151, 84151 Beer-Sheva, Israel
| |
Collapse
|
39
|
Bertelsen RJ, Jensen ET, Ringel-Kulka T. Use of probiotics and prebiotics in infant feeding. Best Pract Res Clin Gastroenterol 2016; 30:39-48. [PMID: 27048895 DOI: 10.1016/j.bpg.2016.01.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 12/21/2015] [Accepted: 01/07/2016] [Indexed: 02/06/2023]
Abstract
Gut colonization by beneficial bacteria in early life is necessary for establishing the gut mucosal barrier, maturation of the immune system and preventing infections with enteric pathogens. Mode of delivery, prematurity, breastfeeding, and use of antibiotics are some of many factors that have been described to influence early life colonization. Dysbiosis, the absence of normal colonization, is associated with many disease conditions. Pre- and probiotics are commonly used as supplementation in infant formula, such as prebiotic oligosaccharides for stimulation of Bifidobacterium growth aiming to mimic the high levels of these commensal bacteria in the gut of breastfed infants. Studies suggest that probiotic supplementation may be beneficial in prevention and management of disease (e.g., reducing the risk of necrotizing enterocolitis in preterm infants and treatment of acute gastroenteritis in children). Although these studies show promising beneficial effects, the long-term risks or health benefits of pre- and probiotic supplementation are not clear.
Collapse
Affiliation(s)
- Randi J Bertelsen
- Department of Clinical Science, University of Bergen, P.O. Box 7804, N-5020 Bergen, Norway; Department of Occupational Medicine, Haukeland University Hospital, P.O. Box 1400, N-5021 Bergen, Norway.
| | - Elizabeth T Jensen
- Wake Forest School of Medicine, Division of Public Health Sciences, Department of Epidemiology & Prevention, Medical Center Boulevard, Winston-Salem, NC 27157, USA; Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7445, USA.
| | - Tamar Ringel-Kulka
- UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, CB# 7445, 404A Rosenau Hall, 421 Pittsboro Street, Chapel Hill, NC 27599-7445, USA.
| |
Collapse
|
40
|
Taipale TJ, Pienihäkkinen K, Isolauri E, Jokela JT, Söderling EM. Bifidobacterium animalis subsp. lactis BB-12 in reducing the risk of infections in early childhood. Pediatr Res 2016; 79:65-9. [PMID: 26372517 DOI: 10.1038/pr.2015.174] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 06/22/2015] [Indexed: 11/09/2022]
Abstract
BACKGROUND Specific probiotic bacteria have proven to be effective in the prevention and treatment of infectious diseases in early life in at-risk populations. The impact of administration of Bifidobacterium animalis subsp. lactis BB-12 (BB-12) on the risk of acute infectious diseases was studied in healthy children. METHODS In this double-blind, placebo-controlled study, 109 1-mo-old infants were assigned randomly to a probiotic group receiving a BB-12-containing tablet (n = 55) or a placebo (n = 54). Test tablets were administered to the infants twice a day (daily dose of BB-12 10 billion colony-forming units) until the age of 2 y with a novel slow-release pacifier or a spoon. Breastfeeding habits, pacifier use, dietary habits, medications, and all signs and symptoms of acute infections were registered in diaries by parents and in questionnaires by trained professionals. RESULTS The infants receiving BB-12 were reported to have experienced fewer respiratory tract infections (RTIs; 87 vs. 100%; risk ratio: 0.87; 95% confidence interval: 0.76, 1.00; P = 0.033) than the controls. No significant differences between the groups were observed in reported gastrointestinal symptoms, otitis media, or fever. The baseline characteristics of the two groups were similar, as was the duration of breastfeeding. CONCLUSION Administration of BB-12 in early childhood may reduce RTIs.
Collapse
Affiliation(s)
| | | | - Erika Isolauri
- Department of Paediatrics, University of Turku, Turku, Finland.,Turku University Hospital, Turku, Finland
| | | | | |
Collapse
|
41
|
Ha GE, Chang OK, Jo SM, Han GS, Park BY, Ham JS, Jeong SG. Identification of Antihypertensive Peptides Derived from Low Molecular Weight Casein Hydrolysates Generated during Fermentation by Bifidobacterium longum KACC 91563. Korean J Food Sci Anim Resour 2015; 35:738-47. [PMID: 26877633 PMCID: PMC4726953 DOI: 10.5851/kosfa.2015.35.6.738] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 06/25/2015] [Accepted: 08/03/2015] [Indexed: 11/13/2022] Open
Abstract
Angiotensin-converting enzyme (ACE) inhibitory activity was evaluated for the low-molecular-weight fraction (<3 kDa) obtained from milk fermentation by Bifidobacterium longum KACC91563. The ACE inhibitory activity in this fraction was 62.3%. The peptides generated from the <3 kDa fraction were identified by liquid chromatography-electrospray ionization quantitative time-of-flight mass spectrometry analysis. Of the 28 peptides identified, 11 and 16 were identified as β-casein (CN) and αs1-CN, respectively. One peptide was identified as κ-CN. Three peptides, YQEPVLGPVRGPFPIIV, QEPVLGPVRGPFPIIV, and GPVRGPFPIIV, from β-CN corresponded to known antihypertensive peptides. We also found 15 peptides that were identified as potential antihypertensive peptides because they included a known antihypertensive peptide fragment. These peptides were as follows: RELEELNVPGEIVE (f1-14), YQEPVLGPVRGPFP (f193-206), EPVLGPVRGPFPIIV (f195-206), PVLGPVRGPFPIIV (f196-206), VLGPVRGPFPIIV (f197-206), and LGPVRGPFPIIV (f198-206) for β-CN; and APSFSDIPNPIGSENSEKTTMPLW (f176-199), SFSDIPNPIGSENSEKT- TMPLW (f178-199), FSDIPNPIGSENSEKTTMPLW (f179-199), SDIPNPIGSENSEKTTMPLW (f180-199), DIPNPIGSENSEKTTMPLW (f181-199), IPNPIGSENSEKTTMPLW (f182-199), PIGSENSEKTTMPLW (f185-199), IGSENSEKTTMPLW (f186-199), and SENSEKTTMPLW (f188-199) for αs1-CN. From these results, B. longum could be used as a starter culture in combination with other lactic acid bacteria in the dairy industry, and/or these peptides could be used in functional food manufacturing as additives for the development of a product with beneficial effects for human health.
Collapse
Affiliation(s)
- Go Eun Ha
- Animal Products Research and Development Division, National Institute of Animal Science, RDA, Jeonju 55365, Korea
| | - Oun Ki Chang
- Animal Products Research and Development Division, National Institute of Animal Science, RDA, Jeonju 55365, Korea; Imported Food Analysis Division, Ministry of Food and Drug Safety, Gwangju 61012, Korea
| | - Su-Mi Jo
- Animal Products Research and Development Division, National Institute of Animal Science, RDA, Jeonju 55365, Korea
| | - Gi-Sung Han
- Animal Products Research and Development Division, National Institute of Animal Science, RDA, Jeonju 55365, Korea
| | - Beom-Young Park
- Animal Products Research and Development Division, National Institute of Animal Science, RDA, Jeonju 55365, Korea
| | - Jun-Sang Ham
- Animal Products Research and Development Division, National Institute of Animal Science, RDA, Jeonju 55365, Korea
| | - Seok-Geun Jeong
- Animal Products Research and Development Division, National Institute of Animal Science, RDA, Jeonju 55365, Korea
| |
Collapse
|
42
|
Khurshid M, Aslam B, Nisar MA, Akbar R, Rahman H, Khan AA, Rasool MH. Bacterial munch for infants: potential pediatric therapeutic interventions of probiotics. Future Microbiol 2015; 10:1881-95. [PMID: 26515509 DOI: 10.2217/fmb.15.102] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Probiotics are viable microorganisms with the capacity to alter the gastrointestinal microbiota of the host. The recent scientific advancements and development of probiotic formulations have rekindled the importance of these clinical interpretations, underlining the starring role of the gut flora in host metabolism, defense and immune regulation. Despite encouraging preliminary results from randomized clinical trials of probiotics for various clinical conditions including irritable bowel syndrome, necrotizing enterocolitis, gastroenteritis, antibiotic-associated diarrhea, infantile colic, and improvement of digestion and immune function, further evidence is needed to determine the reproducibility of the findings and elucidate the underlying mechanisms. In this review, we have considered the postnatal development of gut flora and appraised the role of probiotics in health and disease condition among infants.
Collapse
Affiliation(s)
- Mohsin Khurshid
- College of Allied Health Professionals, Directorate of Medical Sciences, Government College University, Faisalabad, Pakistan.,Department of Microbiology, Government College University, Faisalabad, Pakistan
| | - Bilal Aslam
- Department of Microbiology, Government College University, Faisalabad, Pakistan
| | - Muhammad Atif Nisar
- Department of Microbiology, Government College University, Faisalabad, Pakistan
| | - Rubab Akbar
- Health Biotechnology Division, National Institute for Biotechnology & Genetic Engineering, Faisalabad, Pakistan
| | - Hazir Rahman
- Department of Microbiology, Kohat University of Science & Technology, Kohat, Pakistan
| | - Abdul Arif Khan
- Department of Pharmaceutics, College of Pharmacy, PO Box 2457, King Saud University, Riyadh, 11451, Saudi Arabia
| | | |
Collapse
|
43
|
Abstract
The role of probiotic bacteria in improving human health has been an attractive subject for researchers since the beginning of the 20(th) century. They have been used to control gastro-intestinal infections, to promote immunity and to prevent various diseases (allergies, urogenital infections, etc.). However, the use of beneficial bacteria in the field of dentistry has only recently gained interest. Investigation of the effects of probiotic bacteria on oral health has become an important research subject. These studies are still in the early stages, however results show that probiotic bacteria are effective against tooth caries, periodontal disease, oral mucosal lesions and oral malodour. This review provides information on the effects of probiotics--well-known for their effects on general health, and therefore more widely used in healthcare--on oral and dental health, in order to promote their use/prescription by physicians and patients.
Collapse
Affiliation(s)
- O E Gungor
- 1 Akdeniz University, Faculty of Dentistry, Department of Pediatric Dentistry, 07058 Antalya, Turkey
| | - Z Kirzioglu
- 2 Süleyman Demirel University, Faculty of Dentistry, Department of Pediatric Dentistry, 32260 Isparta, Turkey
| | - M Kivanc
- 3 Anadolu University, Faculty of Science, Department of Biology, 26470 Eskişehir, Turkey
| |
Collapse
|
44
|
Cruchet S, Furnes R, Maruy A, Hebel E, Palacios J, Medina F, Ramirez N, Orsi M, Rondon L, Sdepanian V, Xóchihua L, Ybarra M, Zablah RA. The use of probiotics in pediatric gastroenterology: a review of the literature and recommendations by Latin-American experts. Paediatr Drugs 2015; 17:199-216. [PMID: 25799959 PMCID: PMC4454830 DOI: 10.1007/s40272-015-0124-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE The stability and composition of intestinal flora plays a vital role in human wellbeing throughout life from as early as birth. Over the past 50 years, several studies have been conducted to evaluate the effect of probiotic administration in pediatric gastroenterology. This document aims to provide a recommendation score on probiotic utilization in pediatric gastroenterology, together with a review of current knowledge concerning its benefits, tolerability, and safety. STUDY DESIGN Published literature was selected without study design restriction: clinical guidelines, meta-analyses, randomized controlled trials (RCTs), cohort studies, outcomes research and case-controlled studies were selected using the following MESH-validated terms: probiotics, diarrhea, acute diarrhea, antibiotic-associated diarrhea, traveler's diarrhea, bacterial diarrhea, nosocomial diarrhea, prophylactic diarrhea, Helicobacter pylori infection, colic, infantile colic, necrotizing enterocolitis (NEC), inflammatory bowel disease, constipation, and allergy. Once the validity and the quality of results were evaluated, a recommendation score and level of evidence were assigned for pediatric gastrointestinal-related conditions, according to the updated Evidence-Based Medicine guidelines: 1a for systematic review (SR) of RCTs, 1b for individual RCT, 1c for SR and individual RCT, 2a for SR of cohort studies, 2b for individual cohort studies, 2c for outcomes research, and 3a for SR of case-control studies. RESULTS AND CONCLUSIONS The Latin American Expert group consensus recommends the use of the following probiotics for pediatric gastrointestinal conditions: prevention of acute infectious diarrhea (AID): 1b for Bifidobacterium lactis, Lactobacillus rhamnosus GG (LGG), and L. reuteri; prevention of nosocomial diarrhea: 1 b for B. lactis Bb12, B. bifidum, LGG and Streptococcus thermophiles; treatment of AID: 1a for LGG and S. boulardii, 1b for L. reuteri; prevention of antibiotic-associated diarrhea: 1b for LGG and S. boulardii; prevention of traveler's diarrhea: 1b for S. boulardii; prevention of infantile colic: 1a for L. reuteri DSM 17938; treatment of infantile colic: 1b for L. reuteri DSM 17938; prevention of NEC: 1a for B. breve, mixtures of Bifidobacterium and Streptococcus, LGG, L. acidophilus and L. reuteri DSM 17938; induction and maintenance of remission in ulcerative colitis: 1b for VSL#3; improving symptoms of irritable bowel syndrome: 2c for LGG and VSL#3.
Collapse
Affiliation(s)
- Sylvia Cruchet
- Institute of Nutrition and Food Technology, University of Chile, El Libano 5524, Macul Santiago, Chile,
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Deng K, Wu X, Fuentes C, Su YC, Welti-Chanes J, Paredes-Sabja D, Torres JA. Analysis of Vibrio vulnificus Infection Risk When Consuming Depurated Raw Oysters. J Food Prot 2015; 78:1113-8. [PMID: 26038900 DOI: 10.4315/0362-028x.jfp-14-421] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
A beta Poisson dose-response model for Vibrio vulnificus food poisoning cases leading to septicemia was used to evaluate the effect of depuration at 15 °C on the estimated health risk associated with raw oyster consumption. Statistical variability sources included V. vulnificus level at harvest, time and temperature during harvest and transportation to processing plants, decimal reductions (SV) observed during experimental circulation depuration treatments, refrigerated storage time before consumption, oyster size, and number of oysters per consumption event. Although reaching nondetectable V. vulnificus levels (<30 most probable number per gram) throughout the year and a 3.52 SV were estimated not possible at the 95% confidence level, depuration for 1, 2, 3, and 4 days would reduce the warm season (June through September) risk from 2,669 cases to 558, 93, 38, and 47 cases per 100 million consumption events, respectively. At the 95% confidence level, 47 and 16 h of depuration would reduce the warm and transition season (April through May and October through November) risk, respectively, to 100 cases per 100 million consumption events, which is assumed to be an acceptable risk; 1 case per 100 million events would be the risk when consuming untreated raw oysters in the cold season (December through March).
Collapse
Affiliation(s)
- Kai Deng
- Food Process Engineering Group, Department of Food Science and Technology, Oregon State University, Corvallis, Oregon 97331, USA; Laboratorio de Mecanismos de Patogénesis Bacteriana, Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Santiago, Chile
| | - Xulei Wu
- Seafood Research and Education Center, Oregon State University, Astoria, Oregon 97103, USA
| | - Claudio Fuentes
- Department of Statistics, College of Veterinary Medicine, Oregon State University, Corvallis, Oregon 97331, USA
| | - Yi-Cheng Su
- Seafood Research and Education Center, Oregon State University, Astoria, Oregon 97103, USA
| | - Jorge Welti-Chanes
- Escuela de Ingeniería y Ciencias, Tecnológico de Monterrey, Av. Eugenio Garza Sada 2501 Sur, Colonia Tecnológico, 64849 Monterrey, Nuevo León, México
| | - Daniel Paredes-Sabja
- Laboratorio de Mecanismos de Patogénesis Bacteriana, Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Santiago, Chile; Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University, Corvallis, Oregon 97331, USA.
| | - J Antonio Torres
- Food Process Engineering Group, Department of Food Science and Technology, Oregon State University, Corvallis, Oregon 97331, USA.
| |
Collapse
|
46
|
Ringel-Kulka T, Kotch JB, Jensen ET, Savage E, Weber DJ. Randomized, double-blind, placebo-controlled study of synbiotic yogurt effect on the health of children. J Pediatr 2015; 166:1475-81.e1-3. [PMID: 25841539 DOI: 10.1016/j.jpeds.2015.02.038] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 12/16/2014] [Accepted: 02/10/2015] [Indexed: 12/01/2022]
Abstract
OBJECTIVE To assess the effects of daily consumption of a synbiotic yogurt drink on the health, growth, and quality of life of healthy children 12-48 months of age in out-of-home child care. STUDY DESIGN Healthy children attending child care centers were enrolled in a prospective, double-blind, placebo-controlled clinical trial. The intervention was a yogurt drink containing Streptococcus thermophilus, Lactobacillus bulgaricus, and Bifidobacterium animalis subspecies lactis (BB-12) (5 × 10(9) cfu/100 mL serving), and 1 g of inulin (synbiotic group) vs a similar nonsynbiotic-containing acidified milk drink (placebo group) once daily for 16 weeks. The end points were days of diarrhea, fever, vomiting, symptoms of upper respiratory tract infection, use of antibiotics, physician visits, child care absenteeism, parental work absenteeism, and quality of life (PedsQL 4.0; Mapi Research Trust, Lyon, France). RESULTS Compared with placebo (n = 73), children receiving synbiotic (n = 76) had significantly fewer days of reported fever (1.85 vs 1.95, P < .05), significant improvement in social functioning (P < .035; pre-to-end intervention), and school functioning (P < .045; pre-to-mid intervention). More days with ≥ 3 loose/watery stools were reported in the synbiotic group (P < .05). CONCLUSIONS Daily supplementation of children's diet with yogurt containing probiotic bacteria BB-12 and inulin significantly reduced days of fever and improved social and school functioning. The increased frequency of bowel movements may be explained by an accelerating effect of BB-12 and inulin on intestinal transit. Further research on the possible benefits of synbiotics on children's health is advised. TRIAL REGISTRATION ClinicalTrials.gov: NCT00653705.
Collapse
Affiliation(s)
- Tamar Ringel-Kulka
- Department of Maternal and Child Health, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC.
| | - Jonathan B Kotch
- Department of Maternal and Child Health, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Elizabeth T Jensen
- Department of Maternal and Child Health, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC; Department of Epidemiology, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Eric Savage
- Frank Porter Graham Child Development Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - David J Weber
- Department of Epidemiology, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC; Division of Infectious Diseases, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
47
|
Onubi OJ, Poobalan AS, Dineen B, Marais D, McNeill G. Effects of probiotics on child growth: a systematic review. JOURNAL OF HEALTH, POPULATION, AND NUTRITION 2015; 34:8. [PMID: 26825706 PMCID: PMC5025996 DOI: 10.1186/s41043-015-0010-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 07/23/2014] [Indexed: 06/05/2023]
Abstract
BACKGROUND Child undernutrition has short and long term consequence for both individuals and society. Previous studies show probiotics may promote child growth and have an impact on under-nutrition. METHODS A systematic review of the literature was carried out on three electronic databases to assess evidence. The outcome measured was change in weight or height. A narrative analysis was conducted due to heterogeneity of included studies. RESULTS Twelve studies were included in the review of which ten were randomised controlled trials. A total of 2757 children were included, with 1598 from developing countries. The studies varied in type and quantity of probiotics given, duration of interventions, characteristics of participants, setting and units of outcome measures. Overall, five studies found a positive effect of probiotics on child growth. All five were conducted in developing countries with four studies conducted in mostly under-nourished children and one in well-nourished children. No significant effect on growth was found in the seven studies that were conducted in developed countries. CONCLUSION The limited evidence suggests that probiotics have the potential to improve child growth in developing countries and in under-nourished children. More research is needed to explore this further.
Collapse
Affiliation(s)
- Ojochenemi J Onubi
- Public Health Nutrition Group, Institute of Applied Health Sciences, University of Aberdeen, Polwarth Building, Foresterhill, Aberdeen, AB25 2ZD, UK.
| | - Amudha S Poobalan
- Public Health Nutrition Group, Institute of Applied Health Sciences, University of Aberdeen, Polwarth Building, Foresterhill, Aberdeen, AB25 2ZD, UK.
| | - Brendan Dineen
- Public Health Nutrition Group, Institute of Applied Health Sciences, University of Aberdeen, Polwarth Building, Foresterhill, Aberdeen, AB25 2ZD, UK.
| | - Debbi Marais
- Public Health Nutrition Group, Institute of Applied Health Sciences, University of Aberdeen, Polwarth Building, Foresterhill, Aberdeen, AB25 2ZD, UK.
| | - Geraldine McNeill
- Public Health Nutrition Group, Institute of Applied Health Sciences, University of Aberdeen, Polwarth Building, Foresterhill, Aberdeen, AB25 2ZD, UK.
| |
Collapse
|
48
|
van den Nieuwboer M, Brummer RJ, Guarner F, Morelli L, Cabana M, Claassen E. Safety of probiotics and synbiotics in children under 18 years of age. Benef Microbes 2015; 6:615-30. [PMID: 25809217 DOI: 10.3920/bm2014.0157] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This study aimed to systematically evaluate safety of probiotics and synbiotics in children ageing 0-18 years. This study is the third and final part in a safety trilogy and an update is provided using the most recent available clinical data (2008-2013) by means of the Common Terminology Clinical Adverse Events (CTCAE version 4.0) classification. Safety aspects are represented and related to number of participants per probiotic strain/culture, study duration, dosage, clinical condition and selected afflictions. Analysis of 74 clinical studies indicated that probiotic and/or synbiotic administration in children is safe with regard to the specific evaluated strains, dosages and duration. The population of children include healthy, immune compromised and obese subjects, as well as subjects with intestinal disorders, infections and inflammatory disorders. This study revealed no major safety concerns, as the adverse events (AEs) were unrelated, or not suspected to be related, to the probiotic or synbiotic product. In general the study products were well tolerated. Overall, AEs occurred more frequent in the control arm compared to children receiving probiotics and/or synbiotics. Furthermore, the results indicate inadequate reporting and classification of AEs in the majority of the studies. In addition, generalizability of conclusions are greatly limited by the inconsistent, imprecise and potentially incomplete reporting as well as the variation in probiotic strains, dosages, administration regimes, study populations and reported outcomes.
Collapse
Affiliation(s)
- M van den Nieuwboer
- 1 VU University Amsterdam, Athena Institute, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands
| | - R J Brummer
- 2 School of Health and Medical Sciences, Örebro University, 701 82 Örebro, Sweden
| | - F Guarner
- 3 Digestive System Research Unit, CIBERehd, University Hospital Vall d'Hebron, 08035 Barcelona, Spain
| | - L Morelli
- 4 Istituto di Microbiologia, Università Cattolica S.C., Via Emilia Parmense 84, 29122 Piacenza, Italy
| | - M Cabana
- 5 University of California San Francisco (UCSF), Departments of Pediatrics, Epidemiology and Biostatistics, 3333 California Street, #245, San Francisco, CA 94118, USA
| | - E Claassen
- 1 VU University Amsterdam, Athena Institute, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands.,6 Erasmus Medical Center, Department of Viroscience, P.O. Box 2040, 3000 CA Rotterdam, the Netherlands
| |
Collapse
|
49
|
Probiotic bacteria in infant formula and follow-up formula: Microencapsulation using milk and pea proteins to improve microbiological quality. Food Res Int 2014; 64:567-576. [DOI: 10.1016/j.foodres.2014.07.029] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 06/27/2014] [Accepted: 07/20/2014] [Indexed: 11/18/2022]
|
50
|
Kianifar H, Ahanchian H, Grover Z, Jafari S, Noorbakhsh Z, Khakshour A, Sedaghat M, Kiani M. Synbiotic in the management of infantile colic: a randomised controlled trial. J Paediatr Child Health 2014; 50:801-5. [PMID: 24962875 DOI: 10.1111/jpc.12640] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/06/2014] [Indexed: 01/09/2023]
Abstract
AIM Infant colic is a frequent problem affecting up to 10-30% of infants in first 3 months of life. Results from previous trials have shown that manipulation of gut microbiota can lead to symptomatic improvements. In a randomised clinical trial, we aimed to determine efficacy of synbiotic in reducing average infant crying time at day 7 and day 30 after starting intervention. METHODS Fifty breastfed infants aged 15-120 days with infantile colic randomly assigned to receive either the synbiotic sachet containing 1 billion CFU of: Lactobacillus casei, L. rhamnosus, Streptococcus thermophilus, Bifidobacterium breve, L. acidophilus, B. infantis, L. bulgaricus and fructooligosacharide (Protexin Healthcare, Somerset, UK), or placebo daily for 30 days. Parents were asked to record details of crying times in a symptoms diary. The primary outcome measure was the treatment success (reduction in the daily crying time >50%) and the secondary outcome measure was symptom resolution (reduction in the daily crying time >90%). RESULTS The treatment success was significantly higher in synbiotic group (82.6%) compared with placebo (35.7%) at day 7 (P < 0.005). At day30, treatment success was 87% and 46% in synbiotic and placebo group, respectively (P < 0.01). Symptom resolution was also higher in synbiotic group (39%) compared with placebo (7%) at day 7 (P < 0.03) but not at day 30 (56% vs.36%, P = 0.24). We encountered no complication related to synbiotic use. CONCLUSION This synbiotic (a mixture of seven probiotic strains plus FOS) significantly improved colic symptoms in comparison with placebo.
Collapse
Affiliation(s)
- Hamidreza Kianifar
- Pediatric Gastroenterology, Mashhad University of Medical Sciences, Mashhad, Khorasan, Iran
| | | | | | | | | | | | | | | |
Collapse
|