1
|
Dinleyici EC, Szajewska H, Hojsak I, Gutierrez-Castrellón P, Guarino A, Indrio F, van Goudoever JB, Domellöf M, Shamir R, Savino F, Vandenplas Y. Technical review by the ESPGHAN Special Interest Group on Gut Microbiota and Modifications on the health outcomes of infant formula supplemented with probiotics. J Pediatr Gastroenterol Nutr 2025. [PMID: 40356343 DOI: 10.1002/jpn3.70068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 03/14/2025] [Accepted: 03/31/2025] [Indexed: 05/15/2025]
Abstract
This technical review, one of the five developed by the European Society for Paediatric Gastroenterology Hepatology and Nutrition (ESPGHAN) Special Interest Group on Gut Microbiota and Modifications (SIG-GMM), supports the preparation of a paper on the use of biotic-supplemented formulas, including those containing probiotics. The SIG-GMM conducted this review of studies published before December 31, 2023, to evaluate the clinical outcomes of administering probiotic-supplemented infant formulas to healthy infants (0-12 months). Following the review, all 20 members of the SIG-GMM anonymously assigned scores of 0-9 for each statement related to probiotic-supplemented infant formula. A score ≥6 indicated agreement with a statement, but a statement was rejected if 75% of the members concurred regarding its rejection. The systematic review included 28 studies on the effects of probiotic-supplemented infant formulas. The probiotics studied so far showed no difference compared to the control formula in outcomes such as anthropometric data, gastrointestinal symptoms, stool characteristics, allergy, infections, tolerability and safety. The studies varied in terms of probiotic strains, study designs, and intervention durations. The trials, primarily conducted in Western countries, demonstrated that probiotic-supplemented formulas were well tolerated, with no significant differences in anthropometric parameters and in the growth of infants compared to those fed nonsupplemented formulas in presumed healthy infants. Some evidence suggests potential benefits of probiotic-supplemented formulas in reducing gastrointestinal and respiratory infections, although the findings were inconsistent. This technical review provides the foundation for recommendations on the use of probiotic-supplemented infant formulas in healthy infants.
Collapse
Affiliation(s)
- Ener Cagri Dinleyici
- Department of Pediatrics, Eskisehir Osmangazi University Faculty of Medicine, Eskisehir, Turkey
| | - Hania Szajewska
- Department of Paediatrics, The Medical University of Warsaw, Warsaw, Poland
| | - Iva Hojsak
- Children's Hospital Zagreb, University of Zagreb Medical School, Zagreb, Croatia
| | - Pedro Gutierrez-Castrellón
- Universidad Juarez del Estado de Durango & International Scientific Council for Probiotics S.C., Mexico City, Mexico
| | - Alfredo Guarino
- Pediatric Infectious Disease Unit, Department of Maternal and Child Health, University Hospital "Federico II", Naples, Italy
| | - Flavia Indrio
- Medical School MedTech University of Salento, Lecce, Italy
| | | | - Magnus Domellöf
- Department of Clinical Sciences, Paediatrics, Umeå University, Umeå, Sweden
| | - Raanan Shamir
- Institute of Gastroenterology, Nutrition, and Liver Diseases, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
| | - Francesco Savino
- Ospedale Infantile Regina Margherita di Torino, Città della salute e della Scienza di Torino, Torino, Italy
| | - Yvan Vandenplas
- KidZ Health Castle, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
2
|
Shen SP, Lin HC, Chen JF, Wang HS, Huang YY, Hsia KC, Lin JH, Kuo YW, Li CM, Hsu YC, Tsai SY, Ho HH. Assessment of the safety and gut microbiota modulation ability of an infant formula containing Bifidobacterium animalis ssp. lactis CP-9 or Lactobacillus salivarius AP-32 and the effects of the formula on infant growth outcomes: insights from a four-month clinical study in infants under two months old. BMC Pediatr 2024; 24:840. [PMID: 39731060 DOI: 10.1186/s12887-024-05289-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 11/26/2024] [Indexed: 12/29/2024] Open
Abstract
BACKGROUND Breast milk is a natural treasure for infants, and its microbiota contains a rich array of bacterial species. When breastfeeding is not possible, infant formula with probiotics can be used as a sole source or as a breast milk supplement. The main aim of this study was to evaluate the growth outcomes and tolerance of infants consuming an infant formula containing Bifidobacterium animalis ssp. lactis CP-9 (B. animalis CP-9) or Lactobacillus salivarius AP-32 (L. salivarius AP-32), which were isolated from breast milk and the guts of healthy infants. The safety of these strains in terms of antibiotic resistance and their ability to modulate the gut microbiota were also evaluated. METHODS One hundred eighty healthy infants were included in this study and separated into three groups: the control group, the L. salivarius AP-32 group, and the B. animalis CP-9 group. In this clinical study, adverse events, growth effects, and the incidence of allergies and gastrointestinal disorders in infants consuming infant formula containing B. animalis CP-9 or L. salivarius AP-32 were evaluated. Finally, the impact of the probiotic infant formula on the gut microbiota was elucidated via next-generation sequencing (NGS) analysis. RESULTS The 4-month interventional study revealed that body weight, recumbent length, and head circumference were similar among the three groups. No adverse events related to the intervention were observed. The microbiota composition was more diverse on day 0 and became more uniform by month 4. B. animalis CP-9 and L. salivarius AP-32 were found to be susceptible to streptomycin, tetracycline, erythromycin, clindamycin, chloramphenicol, and ampicillin. CONCLUSIONS The use of infant formula containing B. animalis CP-9 and L. salivarius AP-32 was considered safe and well tolerated, with no adverse events observed during the study. While these strains showed low antibiotic resistance and no immediate concerns related to antibiotic resistance genes, further research is needed to comprehensively assess their long-term safety and efficacy and the potential risk of horizontal gene transfer in broader contexts. TRIAL REGISTRATION The trial was registered with the US Library of Medicine (clinicaltrials.gov) with the number NCT03993301 on 20/06/2019.
Collapse
Affiliation(s)
- Shang-Po Shen
- Division of Neonatology, China Medical University Children's Hospital, Taichung City, Taiwan
| | - Hung-Chih Lin
- Division of Neonatology, China Medical University Children's Hospital, Taichung City, Taiwan
- School of Chinese Medicine, China Medical University, Taichung City, Taiwan
- Department of Pediatrics, Asia University Hospital, Asia University, Taichung City, Taiwan
| | - Jui-Fen Chen
- Research Product Department, R&D Center, Glac Biotech Co., Ltd, Tainan City, Taiwan
| | - Hui-Shan Wang
- Functional R&D Department, R&D Center, Glac Biotech Co., Ltd, Tainan City, Taiwan
| | - Yen-Yu Huang
- Research Product Department, R&D Center, Glac Biotech Co., Ltd, Tainan City, Taiwan
| | - Ko-Chiang Hsia
- Research Product Department, R&D Center, Glac Biotech Co., Ltd, Tainan City, Taiwan
| | - Jia-Hung Lin
- Functional R&D Department, R&D Center, Glac Biotech Co., Ltd, Tainan City, Taiwan
| | - Yi-Wei Kuo
- Functional R&D Department, R&D Center, Glac Biotech Co., Ltd, Tainan City, Taiwan
| | - Ching-Min Li
- Research Product Department, R&D Center, Glac Biotech Co., Ltd, Tainan City, Taiwan
| | - Yu-Chieh Hsu
- Research Product Department, R&D Center, Glac Biotech Co., Ltd, Tainan City, Taiwan
| | - Shin-Yu Tsai
- Research Product Department, R&D Center, Glac Biotech Co., Ltd, Tainan City, Taiwan
| | - Hsieh-Hsun Ho
- Research Product Department, R&D Center, Glac Biotech Co., Ltd, Tainan City, Taiwan.
- Functional R&D Department, R&D Center, Glac Biotech Co., Ltd, Tainan City, Taiwan.
| |
Collapse
|
3
|
Berseth CL, Yeiser M, Harris CL, Kinnaman JN, Lappin V, Wampler JL, Zhuang W, Vanderhoof J. Infant formula with added Lacticaseibacillus rhamnosus GG supported adequate growth and was well tolerated in healthy term infants: a randomized controlled trial. Front Pediatr 2024; 12:1456607. [PMID: 39507493 PMCID: PMC11538007 DOI: 10.3389/fped.2024.1456607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/24/2024] [Indexed: 11/08/2024] Open
Abstract
Introduction Lacticaseibacillus rhamnosus GG (LGG) is a well-studied probiotic with a history of safe use. Methods In this double-blind, prospective study, growth and tolerance were evaluated in healthy term infants randomized to: marketed, routine intact cow's milk protein-based formula (Control, n = 172) or a similar investigational formula with added LGG (INV-LGG, n = 179; 106 CFU LGG®/g powder) from 14 to 120 days of age. Anthropometrics, stool characteristics, fussiness, and gassiness were evaluated through Day 120. Medically confirmed adverse events were recorded throughout the study period. The primary outcome was rate of weight gain from Day 14-120. Results Of 351 infants enrolled, 275 completed (Control, n = 131; INV-LGG, n = 144). No significant group differences in rate of weight gain from Day 14-120 were detected. Study formula acceptance and tolerance was good with no significant differences in study discontinuation due to study formula or parent-reported gassiness, stool frequency, or stool consistency; however mean fussiness relative to normal was significantly lower for INV-LGG vs Control at Days 60 and 90. Discussion In healthy term infants, a routine intact cow's milk protein-based formula with added LGG supported adequate growth and was well tolerated. Further studies are needed to evaluate potential benefits for fussiness and efficacy outcomes. Clinical Trial Registration Clinicaltrials.gov, identifier (NCT01897922).
Collapse
Affiliation(s)
- Carol Lynn Berseth
- Medical Sciences, Reckitt | Mead Johnson Nutrition, Evansville, IN, United States
| | | | - Cheryl L. Harris
- Medical Sciences, Reckitt | Mead Johnson Nutrition, Evansville, IN, United States
| | - Jennifer N. Kinnaman
- Nutrition Sciences, Reckitt | Mead Johnson Nutrition, Evansville, IN, United States
| | - Victoria Lappin
- Clinical Research, Research & Development, Reckitt, Hull, United Kingdom
| | - Jennifer L. Wampler
- Medical Sciences, Reckitt | Mead Johnson Nutrition, Evansville, IN, United States
| | - Weihong Zhuang
- Medical Sciences, Reckitt | Mead Johnson Nutrition, Evansville, IN, United States
| | - Jon Vanderhoof
- Gastroenterology, Boston Children’s Hospital, Boston, MA, United States
| |
Collapse
|
4
|
Catassi G, Aloi M, Giorgio V, Gasbarrini A, Cammarota G, Ianiro G. The Role of Diet and Nutritional Interventions for the Infant Gut Microbiome. Nutrients 2024; 16:400. [PMID: 38337684 PMCID: PMC10857663 DOI: 10.3390/nu16030400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/14/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
The infant gut microbiome plays a key role in the healthy development of the human organism and appears to be influenced by dietary practices through multiple pathways. First, maternal diet during pregnancy and infant nutrition significantly influence the infant gut microbiota. Moreover, breastfeeding fosters the proliferation of beneficial bacteria, while formula feeding increases microbial diversity. The timing of introducing solid foods also influences gut microbiota composition. In preterm infants the gut microbiota development is influenced by multiple factors, including the time since birth and the intake of breast milk, and interventions such as probiotics and prebiotics supplementation show promising results in reducing morbidity and mortality in this population. These findings underscore the need for future research to understand the long-term health impacts of these interventions and for further strategies to enrich the gut microbiome of formula-fed and preterm infants.
Collapse
Affiliation(s)
- Giulia Catassi
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (G.C.); (A.G.); (G.C.)
- Pediatric Gastroenterology and Liver Unit, Sapienza University of Rome, Umberto I Hospital, 00161 Rome, Italy;
| | - Marina Aloi
- Pediatric Gastroenterology and Liver Unit, Sapienza University of Rome, Umberto I Hospital, 00161 Rome, Italy;
| | - Valentina Giorgio
- Department of Woman and Child Health and Public Health, UOC Pediatria, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy;
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (G.C.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Giovanni Cammarota
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (G.C.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Gianluca Ianiro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (G.C.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|
5
|
De Bernardo G, D’Urso G, Spadarella S, Giordano M, Leone G, Casapullo A. Analysis of the Fecal Metabolomic Profile in Breast vs. Different Formula Milk Feeding in Late Preterm Infants. Metabolites 2024; 14:72. [PMID: 38276307 PMCID: PMC10820811 DOI: 10.3390/metabo14010072] [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: 12/20/2023] [Revised: 01/12/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
Human milk is the gold standard for infant nutrition, but when it is not available or insufficient to satisfy the needs of the infant, formula milk is proposed as an effective substitute. A prospective observational cohort study was conducted on late preterm infants fed with breast and two different formula milks. On this basis, they were divided into three groups: group FMPB (fed with formula + postbiotic), group FM (fed with standard formula), and group BM (breastfed). Stool samples for a metabolomic study were collected at T0 (5-7 days after birth), T1 (30 days of life), and T2 (90 days of life), giving rise to 74 samples analyzed via liquid chromatography coupled with high-resolution mass spectrometry. The T0, T1, and T2 LC-MS raw data were processed for Partial Least Square Discriminant Analysis (PLS-DA), followed by a statistical analysis. This preliminary study highlighted a good overlapping between the fecal metabolome of breast and substitute feeding systems, confirming the efficacy of the formula preparations as breast milk substitutes. Moreover, several similarities were also detected between the FMPB and BM metabolome, highlighting that the addition of a postbiotic to standard formula milk could be more effective and considered a better alternative to breast milk.
Collapse
Affiliation(s)
- Giuseppe De Bernardo
- Division of Pediatrics Neonatology and NICU, Ospedale Buon Consiglio Fatebenefratelli, 80123 Naples, Italy; (S.S.); (G.L.)
| | - Gilda D’Urso
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy;
| | - Simona Spadarella
- Division of Pediatrics Neonatology and NICU, Ospedale Buon Consiglio Fatebenefratelli, 80123 Naples, Italy; (S.S.); (G.L.)
| | - Maurizio Giordano
- Department of Clinical Medicine and Surgery, Federico II University, 80138 Naples, Italy;
| | - Giuseppina Leone
- Division of Pediatrics Neonatology and NICU, Ospedale Buon Consiglio Fatebenefratelli, 80123 Naples, Italy; (S.S.); (G.L.)
| | - Agostino Casapullo
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy;
| |
Collapse
|
6
|
Eor JY, Lee CS, Moon SH, Cheon JY, Pathiraja D, Park B, Shin MJ, Kim JY, Kim S, Noh Y, Kim Y, Choi IG, Kim SH. Effect of Probiotic-Fortified Infant Formula on Infant Gut Health and Microbiota Modulation. Food Sci Anim Resour 2023; 43:659-673. [PMID: 37484007 PMCID: PMC10359846 DOI: 10.5851/kosfa.2023.e26] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/09/2023] [Accepted: 06/09/2023] [Indexed: 07/25/2023] Open
Abstract
Compared to infant formula, breast milk is the best source of nutrition for infants; it not only improves the neonatal intestinal function, but also regulates the immune system and gut microbiota composition. However, probiotic-fortified infant formula may further enhance the infant gut environment by overcoming the limitations of traditional infant formula. We investigated the probiotic formula administration for one month by comparing 118 Korean infants into the following three groups: infants in each group fed with breast milk (50), probiotic formula (35), or placebo formula-fed group (33). Probiotic formula improved stool consistency and defecation frequency compared to placebo formula-fed group. The probiotic formula helped maintaining the level of secretory immunoglobulin A (sIgA), which had remarkably decreased over time in placebo formula-fed infants (compared to weeks 0 and 4). Moreover, probiotic formula decreased the acidity of stool and considerably increased the butyrate concentration. Furthermore, the fecal microbiota of each group was evaluated at weeks 0 and 4. The microbial composition was distinct between each groups, and the abundance of health-promoting bacteria increased in the probiotic formula compared to the placebo formula-fed group. In summary, supplementation of probiotic infant formula can help optimize the infant gut environment, microbial composition, and metabolic activity of the microbiota, mimicking those of breast milk.
Collapse
Affiliation(s)
- Ju Young Eor
- College of Life Sciences and
Biotechnology, Korea University, Seoul 02841, Korea
| | - Chul Sang Lee
- College of Life Sciences and
Biotechnology, Korea University, Seoul 02841, Korea
- Institute of Life Sciences and Natural
Resources, Korea University, Seoul 02841, Korea
| | - Sung Ho Moon
- College of Life Sciences and
Biotechnology, Korea University, Seoul 02841, Korea
| | - Ju Young Cheon
- College of Life Sciences and
Biotechnology, Korea University, Seoul 02841, Korea
| | - Duleepa Pathiraja
- College of Life Sciences and
Biotechnology, Korea University, Seoul 02841, Korea
| | - Byeonghyeok Park
- College of Life Sciences and
Biotechnology, Korea University, Seoul 02841, Korea
| | - Min Jae Shin
- College of Life Sciences and
Biotechnology, Korea University, Seoul 02841, Korea
| | - Jae-Young Kim
- College of Life Sciences and
Biotechnology, Korea University, Seoul 02841, Korea
- Institute of Life Sciences and Natural
Resources, Korea University, Seoul 02841, Korea
| | | | | | | | - In-Geol Choi
- College of Life Sciences and
Biotechnology, Korea University, Seoul 02841, Korea
- Institute of Life Sciences and Natural
Resources, Korea University, Seoul 02841, Korea
| | - Sae Hun Kim
- College of Life Sciences and
Biotechnology, Korea University, Seoul 02841, Korea
- Institute of Life Sciences and Natural
Resources, Korea University, Seoul 02841, Korea
| |
Collapse
|
7
|
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: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [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
|
8
|
Xavier-Santos D, Scharlack NK, Pena FDL, Antunes AEC. Effects of Lacticaseibacillus rhamnosus GG supplementation, via food and non-food matrices, on children's health promotion: A scoping review. Food Res Int 2022; 158:111518. [PMID: 35840226 DOI: 10.1016/j.foodres.2022.111518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 06/11/2022] [Accepted: 06/13/2022] [Indexed: 11/04/2022]
Abstract
The literature considers children both a risk group for administering probiotic strains and one of the populations that can most benefit from it. Due to the health benefits associated to probiotic supplementation, this scope review sought to formulate a critical evaluation of how Lacticaseibacillus rhamnosus GG, carried in food and non-food matrices, and experimental design may affect the health promotion of infants and children. In this study, a literature search was conducted in three scientific databases: PubMed, Web of Science, and SciELO to retrieve research, published in English or Spanish, which administered L. rhamnosus GG to infants and children with any disease or in eutrophic condition. Three reviewers with an expert supervision screened 540 articles, published between 2001 and 2022, which were retrieved from the databases. The data extracted was compiled and shown in this scoping review. In total, was included, after criteria observation, 44 articles in this review. Intestinal disorders were the most frequent outcome in these studies (36.4%) and capsules, the most common vehicle for administering the probiotic strain (40.9%). Probiotic strain dose ranged from 105 to 1012 cfu/dose of L. rhamnosus GG and intervention length extended from one to more than 6 months. Food matrix showed health effects in 57.1% of the clinical trials and non-food matrix 46.7%, which indicates that the health-promoting effect of the probiotic GG strain may be equivalent between the two forms of delivery. However, the highly heterogeneous experimental designs prevent further analysis and a systematic review and meta-analysis is recommended to address just the outcomes of studies and achieve data homogeneity in order to determine which vehicle is the most suitable for health promoting.
Collapse
Affiliation(s)
- Douglas Xavier-Santos
- School of Applied Sciences (FCA), State University of Campinas, 300 Pedro Zaccaria St, 13484-350 Limeira, SP, Brazil
| | - Nayara Kastem Scharlack
- School of Applied Sciences (FCA), State University of Campinas, 300 Pedro Zaccaria St, 13484-350 Limeira, SP, Brazil
| | - Fabíola de Lima Pena
- School of Applied Sciences (FCA), State University of Campinas, 300 Pedro Zaccaria St, 13484-350 Limeira, SP, Brazil
| | | |
Collapse
|
9
|
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
|
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
|
Qi M, Tan B, Wang J, Liao S, Deng Y, Ji P, Song T, Zha A, Yin Y. The microbiota-gut-brain axis: A novel nutritional therapeutic target for growth retardation. Crit Rev Food Sci Nutr 2021; 62:4867-4892. [PMID: 33523720 DOI: 10.1080/10408398.2021.1879004] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Growth retardation (GR), which commonly occurs in childhood, is a major health concern globally. However, the specific mechanism remains unclear. It has been increasingly recognized that changes in the gut microbiota may lead to GR through affecting the microbiota-gut-brain axis. Microbiota interacts with multiple factors such as birth to affect the growth of individuals. Microbiota communicates with the nerve system through chemical signaling (direct entry into the circulation system or stimulation of enteroendocrine cells) and nervous signaling (interaction with enteric nerve system and vagus nerve), which modulates appetite and immune response. Besides, they may also influence the function of enteric glial cells or lymphocytes and levels of systemic inflammatory cytokines. Environmental stress may cause leaky gut through perturbing the hypothalamic-pituitary-adrenal axis to further result in GR. Nutritional therapies involving probiotics and pre-/postbiotics are being investigated for helping the patients to overcome GR. In this review, we summarize the role of microbiota in GR with human and animal models. Then, existing and potential regulatory mechanisms are reviewed, especially the effect of microbiota-gut-brain axis. Finally, we propose nutritional therapeutic strategies for GR by the intervention of microbiota-gut-brain axis, which may provide novel perspectives for the treatment of GR in humans and animals.
Collapse
Affiliation(s)
- Ming Qi
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China.,College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Bie Tan
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China.,College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
| | - Jing Wang
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| | - Simeng Liao
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yuankun Deng
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China.,College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
| | - Peng Ji
- Department of Nutrition, University of California, Davis, California, USA
| | - Tongxing Song
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Andong Zha
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yulong Yin
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China.,College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
| |
Collapse
|
12
|
Westerik N, Nelson A, Wacoo AP, Sybesma W, Kort R. A Comparative Interrupted Times Series on the Health Impact of Probiotic Yogurt Consumption Among School Children From Three to Six Years Old in Southwest Uganda. Front Nutr 2020; 7:574792. [PMID: 33363193 PMCID: PMC7756026 DOI: 10.3389/fnut.2020.574792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 11/19/2020] [Indexed: 12/12/2022] Open
Abstract
Introduction: Following a school milk feeding program in Southwest Uganda, we initiated a probiotic yogurt school feeding program in the same region in 2018. In order to investigate the potential health benefits from probiotic yogurt we conducted an observational study, where we compared the effect of the consumption of locally produced probiotic yogurt containing Lactobacillus rhamnosus yoba 2012 to milk in pre-primary schoolchildren from different schools on the occurrence of respiratory tract infections (common cold) and skin infections (e.g., tinea capitis). Method: A comparative interrupted time series over a period of 3 weeks of baseline followed by 9 weeks of 100 ml of probiotic yogurt or milk consumption for 5 days per week. In total 584 children attending five different schools were followed during consumption of probiotic yogurt and 532 children attending five other schools during consumption of milk. Incidences of respiratory tract infection symptoms and skin infection symptoms, changes in anthropometric indicators and absenteeism were recorded. Results: Over the course of the study period the incidence rate for common cold symptoms decreased faster in the yogurt group than in the milk group (p = 0.09) resulting in a final RR of 0.85 (95% CI: 0.5-1.4) at the end of the observational period. The incidence rate of skin infection related symptoms also reduced faster in the yogurt group compared to the milk group (p < 0.0001) resulting in a relative risk factor (RR) of 0.6 (CI: 0.4-0.9) at the end of the observational period. Anthropometric indicators and level of absenteeism did not show significant differences between yogurt and milk. Conclusion: Notwithstanding the observed positive trend and effect of probiotic yogurt on the incidences of common cold and skin infections, respectively, we consider the results of this comparative interrupted time series inconclusive due to differences in the recorded health parameters between the probiotic yogurt and milk control groups at base line, and fluctuations over the course of the intervention period. An improved study design, with more uniform study groups, a longer intervention period and a third control group without yogurt or milk is required to draw definitive conclusions.
Collapse
Affiliation(s)
- Nieke Westerik
- Yoba for Life Foundation, Amsterdam, Netherlands
- Department of Molecular Cell Biology, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | | | - Alex Paul Wacoo
- Yoba for Life Foundation, Amsterdam, Netherlands
- Department of Molecular Cell Biology, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Department of Food Technology and Nutrition, School of Food Technology Nutrition and Bioengineering, College of Agricultural and Environmental Sciences, Makerere University, Kampala, Uganda
| | | | - Remco Kort
- Yoba for Life Foundation, Amsterdam, Netherlands
- Department of Molecular Cell Biology, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
13
|
Laomongkholchaisri P, Teanpaisan R, Wonglapsuwan M, Piwat S. Impact of Potential Probiotic Lactobacillus Strains on Host Growth and Development in a Drosophila melanogaster Model. Probiotics Antimicrob Proteins 2020; 13:390-397. [PMID: 32875521 DOI: 10.1007/s12602-020-09705-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2020] [Indexed: 11/24/2022]
Abstract
Lactobacillus paracasei SD1, Lactobacillus rhamnosus SD11, and Lactobacillus gasseri SD12 were proposed as potential probiotics for oral health. However, the effects of them on host physiology are still unknown. This study aimed to select strains that can promote host growth and development in monocolonized Drosophila model compared with axenic and the commercial Lactobacillus rhamnosus GG-treated flies. The morphogenesis and growth of axenic Drosophila melanogaster were assessed from embryo to adult stage when provided with each probiotic strain-supplemented food. The colonization and persistence of probiotic in fly gut were also evaluated. The results indicated that axenic condition caused the lowest adult weight and emergence rate. All probiotic groups had higher weight than axenic group. Lact. rhamnosus SD11 group presented high adult emergence rate equivalent to Lact. rhamnosus GG group, which is significantly higher than the others. However, Lact. gasseri SD12 group had significantly lower adult weight and emergence rate when compared with Lact. rhamnosus GG group. The gut probiotics levels were rapidly increased within the first day after receiving probiotics. After probiotic cessation, their number in gut decreased and was maintained at low level, except for Lact. gasseri SD12, which completely vanished since day one. In conclusion, Lact. paracasei SD1, Lact. rhamnosus SD11, and Lact. rhamnosus GG can affect morphogenesis and weight of flies when fed since immature stage and have short period gut colonization. The findings of this study could possibly imply comparable health-promoting effects between Lact. paracasei SD1 and Lact. rhamnosus SD11 to commercial strain in Drosophila model.
Collapse
Affiliation(s)
- Pasaraporn Laomongkholchaisri
- Department of Preventive Dentistry, Faculty of Dentistry, Prince of Songkla University, Hat-Yai, Thailand.,Common Oral Diseases and Epidemiology Research Center, Faculty of Dentistry, Prince of Songkla University, Hat-Yai, Thailand
| | - Rawee Teanpaisan
- Common Oral Diseases and Epidemiology Research Center, Faculty of Dentistry, Prince of Songkla University, Hat-Yai, Thailand.,Department of Stomatology, Faculty of Dentistry, Prince of Songkla University, Hat-Yai, Thailand
| | - Monwadee Wonglapsuwan
- Department of Molecular Biotechnology and Bioinformatics, Faculty of Science, Prince of Songkla University, Hat-Yai, Thailand
| | - Supatcharin Piwat
- Department of Preventive Dentistry, Faculty of Dentistry, Prince of Songkla University, Hat-Yai, Thailand. .,Common Oral Diseases and Epidemiology Research Center, Faculty of Dentistry, Prince of Songkla University, Hat-Yai, Thailand.
| |
Collapse
|
14
|
Gaukroger CH, Stewart CJ, Edwards SA, Walshaw J, Adams IP, Kyriazakis I. Changes in Faecal Microbiota Profiles Associated With Performance and Birthweight of Piglets. Front Microbiol 2020; 11:917. [PMID: 32595608 PMCID: PMC7300224 DOI: 10.3389/fmicb.2020.00917] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 04/17/2020] [Indexed: 12/22/2022] Open
Abstract
The gastrointestinal tract microbiota interacts with the host to modulate metabolic phenotype. This interaction could provide insights into why some low birthweight pigs can exhibit compensatory growth whilst others remain stunted. This study aimed to identify microbiota markers associated with birthweight [low birthweight (n = 13) or normal birthweight pigs (n = 13)] and performance ["good" or "poor" average daily gain (ADG) class]. Furthermore, the study determined whether the taxonomic markers were longitudinal, or time point specific in their ability to identify low birthweight pigs who could exhibit compensatory growth. Faecal samples were collected and liveweight recorded at 10 different time points from birth to 56 days of age. No consistent associations between birthweight, performance and gut microbiota were found across all time points. However, there was a significant (P < 0.05) effect of birthweight on microbiota richness at 21, 27, 32 and 56 days of age. Significant differences (P < 0.05) in genera abundance according to birthweight and performance were also identified. Low birthweight pigs had a significantly (P < 0.05) lower abundance of Ruminococcaceae UCG-005, but a significantly (P < 0.05) higher abundance of Ruminococcaceae UCG-014 on days 21 and 32, respectively. Piglets classified as having a "good" ADG class had a significantly (P < 0.05) higher abundance of Lactobacillus, unclassified Prevotellaceae and Ruminococcaceae UCG-005 on days 4, 8 and 14, respectively. Furthermore, Ruminococcaceae UCG-005 was significantly more abundant at 14 days of age in normal birthweight pigs with a "good" ADG class compared to those classified as "poor." The results of this study indicate that there are time point-specific differences in the microbiota associated with birthweight and performance, corresponding to the period in which solid feed intake first occurs. Identifying early-life microbiota markers associated with performance emphasises the importance of the neonatal phase when considering intervention strategies aimed at promoting performance.
Collapse
Affiliation(s)
- Clare H. Gaukroger
- Agriculture, School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Christopher J. Stewart
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Sandra A. Edwards
- Agriculture, School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | - Ian P. Adams
- Fera Science Limited, York, United Kingdom
- Institute for Agri-Food Research and Innovation, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Ilias Kyriazakis
- School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| |
Collapse
|
15
|
Analysis of immune, microbiota and metabolome maturation in infants in a clinical trial of Lactobacillus paracasei CBA L74-fermented formula. Nat Commun 2020; 11:2703. [PMID: 32483147 PMCID: PMC7264213 DOI: 10.1038/s41467-020-16582-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 05/05/2020] [Indexed: 12/15/2022] Open
Abstract
Mother’s milk is the best choice for infants nutrition, however when it is not available or insufficient to satisfy the needs of the infant, formula is proposed as an effective substitute. Here, we report the results of a randomized controlled clinical trial (NCT03637894) designed to evaluate the effects of two different dietary regimens (standard formula and Lactobacillus paracasei CBA L74-fermented formula) versus breastfeeding (reference group) on immune defense mechanisms (primary endpoint: secretory IgA, antimicrobial peptides), the microbiota and its metabolome (secondary outcomes), in healthy full term infants according to the type of delivery (n = 13/group). We show that the fermented formula, safe and well tolerated, induces an increase in secretory IgA (but not in antimicrobial peptides) and reduces the diversity of the microbiota, similarly, but not as much as, breastmilk. Metabolome analysis allowed us to distinguish subjects based on their dietary regimen and mode of delivery. Together, these results suggest that a fermented formula favors the maturation of the immune system, microbiota and metabolome. Milk breastfeeding and prebiotic-supplemented formulas have varying effects on the infant gut microbiome. Here, in a randomized controlled clinical trial, the authors investigate the effects of a Lactobacillus paracasei-fermented formula on the immune defense mechanisms, microbiota and its metabolome in full term infants.
Collapse
|
16
|
Karkhaneh M, Fraser L, Jou H, Vohra S. Effectiveness of probiotics in infantile colic: A rapid review. Paediatr Child Health 2020; 25:149-159. [PMID: 32296276 DOI: 10.1093/pch/pxz007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 07/31/2018] [Indexed: 01/20/2023] Open
Abstract
Background Infantile colic (IC) is a troubling condition with limited treatment options for young infants. This rapid review aims to synthesize the evidence for probiotics in the treatment and prevention of IC in healthy term infants. Methods We searched in MEDLINE, EMBASE, CINAHL, Cochrane Central Register of Controlled Trials and Cochrane Database of Systematic Reviews for systematic reviews (SRs), and randomized control trials (RCTs) published between January 1, 2000 and July 11, 2018. Trials were included if they recruited healthy full-term infants who received probiotics for treatment or prevention of colic. The quality of evidence was assessed using GRADE criteria. As supplementary information, the safety of probiotics in infants was searched within the reviewed studies and other recent publications. Results We identified four SRs and meta-analyses that included six RCTs, and found an additional three RCTs evaluating probiotics for the treatment of IC. One SR and six RCTs were identified for prevention of IC; four of the RCTs were included in the SR and two were published later. The probiotic Lactobacillus reuteri was used in the majority (five of eight) of treatment trials, and was found to significantly reduce crying in colicky breast-fed infants compared to placebo. Only two of the six prevention trials showed a significant decrease in crying time compared to placebo, although another two trials showed other benefits of probiotics, including reduced use of medications (simethicone and cimetropium bromide) and physician visits. No adverse events were identified in the included studies; other research suggests probiotics are generally safe in healthy children. Conclusion This rapid review identified limited but favourable evidence of benefit of using probiotics for the treatment of IC in full-term breast-fed infants. While routine use of probiotics for treating or preventing IC cannot yet be recommended, it can be an option to manage IC.
Collapse
Affiliation(s)
- Mohammad Karkhaneh
- Department of Pediatrics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta
| | - Lexa Fraser
- Department of Pediatrics & Child Health, University of Manitoba, Winnipeg, Manitoba
| | - Hsing Jou
- Department of Pediatrics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta
| | - Sunita Vohra
- Department of Pediatrics, Medicine, and Psychiatry, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta
| |
Collapse
|
17
|
Microbiome Composition in Pediatric Populations from Birth to Adolescence: Impact of Diet and Prebiotic and Probiotic Interventions. Dig Dis Sci 2020; 65:706-722. [PMID: 32002758 PMCID: PMC7046124 DOI: 10.1007/s10620-020-06092-x] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Diet is a key regulator of microbiome structure and function across the lifespan. Microbial colonization in the first year of life has been actively researched; however, studies during childhood are sparse. Herein, the impact of dietary intake and pre- and probiotic interventions on microbiome composition of healthy infants and children from birth to adolescence is discussed. The microbiome of breastfed infants has lower microbial diversity and richness, higher Proteobacteria, and lower Bacteroidetes and Firmicutes than those formula-fed. As children consume more complex diets, associations between dietary patterns and the microbiota emerge. Like adults, the microbiota of children consuming a Western-style diet is associated with greater Bacteroidaceae and Ruminococcaceae and lower Prevotellaceae. Dietary fibers and pre- or/and probiotics have been tested to modulate the gut microbiota in early life. Human milk oligosaccharides and prebiotics added to infant formula are bifidogenic and decrease pathogens. In children, prebiotics, such as inulin, increase Bifidobacterium abundance and dietary fibers reduce fecal pH and increase alpha diversity and calcium absorption. Probiotics have been administered to the mother during pregnancy and breastfeeding or directly to the infant/child. Findings on maternal probiotic administration on bacterial taxa are inconsistent. When given directly to the infant/child, some changes in individual taxa are observed, but rarely is overall alpha or beta diversity affected. Cesarean-delivered infants appear to benefit to a greater degree than those born vaginally. Infancy and childhood represent an opportunity to beneficially manipulate the microbiome through dietary or prebiotic interventions, which has the potential to affect both short- and long-term health outcomes.
Collapse
|
18
|
Maldonado J, Gil-Campos M, Maldonado-Lobón JA, Benavides MR, Flores-Rojas K, Jaldo R, Jiménez Del Barco I, Bolívar V, Valero AD, Prados E, Peñalver I, Olivares M. Evaluation of the safety, tolerance and efficacy of 1-year consumption of infant formula supplemented with Lactobacillus fermentum CECT5716 Lc40 or Bifidobacterium breve CECT7263: a randomized controlled trial. BMC Pediatr 2019; 19:361. [PMID: 31630683 PMCID: PMC6802336 DOI: 10.1186/s12887-019-1753-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 09/27/2019] [Indexed: 01/18/2023] Open
Abstract
Background The microorganism present in breast milk, added to other factors, determine the colonization of infants. The objective of the present study is to evaluate the safety, tolerance and effects of the consumption of a milk formula during the first year of life that is supplemented with L. fermentum CECT5716 or Bifidobacterium breve CECT7263, two strains originally isolated from breast milk. Methods A randomized, double blind, controlled, parallel group study including healthy, formula-fed infants was conducted. Two hundred and thirty-six 1-month-old infants were selected and randomly divided into three study groups according to a randomization list. Infants in the control group received a standard powdered infant formula until 12 months of age. Infants in the probiotic groups received the same infant formula but supplemented with L. fermentum CECT5716 Lc40 or B. breve CECT7263. Main outcome was weigh-gain of infants as safety marker. Results One hundred and eighty-nine infants completed the eleven months of intervention (61 in control group, 65 in Lf group and 63 in Bb group). The growth of infants in the three groups was consistent with standards. No significant differences were observed in the main outcome, weight-gain (Control group: 5.77 Kg ± 0.95, Lf group: 5.77 Kg ± 1.31, Bb group: 5.58 Kg ± 1.10; p = 0.527). The three milk formulae were well tolerated, and no adverse effects were related to the consumption of any of the formula. Infants receiving B. breve CECT7263 had a 1.7 times lower risk of crying than the control group (OR = 0.569, CI 95% 0.568–0.571; p = 0.001). On the other hand, the incidence of diarrhoea in infants receiving the formula supplemented with L. fermentum CECT5716 was a 44% lower than in infants receiving the control formula (p = 0.014). The consumption of this Lactobacillus strain also reduced the duration of diarrhoea by 2.5 days versus control group (p = 0.044). Conclusions The addition of L. fermentum CECT5716 Lc40 or B. breve CECT7263, two probiotic strains naturally found in breast milk, to infant formulae is safe and induces beneficial effects on the health of infants. Trial registration The trial was retrospectively registered in the US Library of Medicine (www.clinicaltrial.gov) with the number NCT03204630. Registered 11 August 2016.
Collapse
Affiliation(s)
- J Maldonado
- Pediatric Unit, University Hospital Virgen de las Nieves, Granada, Spain.,Pediatric Department, University of Granada, Granada, Spain.,Biosanitary Research Institute (IBS), Granada, Spain.,Maternal and Child Health and Development Network (SAMID), Health Institute Carlos III, Madrid, Spain
| | - M Gil-Campos
- Unit of Metabolism and Pediatric Research (IMIBIC), Reina Sofia University Hospital, University of Córdoba, Cordoba, Spain.,CIBEROBN, Cordoba, Spain
| | | | - M R Benavides
- Pediatric Clinic Roquetas, Roquetas de Mar, Almería, Spain
| | - K Flores-Rojas
- Unit of Metabolism and Pediatric Research (IMIBIC), Reina Sofia University Hospital, University of Córdoba, Cordoba, Spain
| | - R Jaldo
- Andalusian Health Service, Andalusia, Spain
| | | | - V Bolívar
- Andalusian Health Service, Andalusia, Spain
| | - A D Valero
- Biosearch Life, Research Department, Granada, Spain
| | - E Prados
- Andalusian Health Service, Andalusia, Spain
| | - I Peñalver
- Andalusian Health Service, Andalusia, Spain
| | - M Olivares
- Biosearch Life, Research Department, Granada, Spain.
| |
Collapse
|
19
|
Abstract
Lactobacillus rhamnosus GG (LGG) was the first strain belonging to the genus Lactobacillus to be patented in 1989 thanks to its ability to survive and to proliferate at gastric acid pH and in medium containing bile, and to adhere to enterocytes. Furthermore LGG is able to produces both a biofilm that can mechanically protect the mucosa, and different soluble factors beneficial to the gut by enhancing intestinal crypt survival, diminishing apoptosis of the intestinal epithelium, and preserving cytoskeletal integrity. Moreover LGG thanks to its lectin-like protein 1 and 2 inhibits some pathogens such as Salmonella species. Finally LGG is able to promote type 1 immune-responsiveness by reducing the expression of several activation and inflammation markers on monocytes and by increasing the production of interleukin-10, interleukin-12 and tumor necrosis factor-α in macrophages. A large number of research data on Lactobacillus GG is the basis for the use of this probiotic for human health. In this review we have considered predominantly randomized controlled trials, meta-analysis, Cochrane Review, guide lines of Scientific Societies and anyway studies whose results were evaluated by means of relative risk, odds ratio, weighted mean difference 95% confidence interval. The effectiveness of LGG in gastrointestinal infections and diarrhea, antibiotic and Clostridium difficile associated diarrhea, irritable bowel syndrome, inflammatory bowel disease, respiratory tract infections, allergy, cardiovascular diseases, nonalcoholic fatty liver disease, nonalcoholic steatohepatitis, cystic fibrosis, cancer, elderly end sport were analyzed.
Collapse
|
20
|
Nguyen TTB, Chung HJ, Kim HJ, Hong ST. Establishment of an ideal gut microbiota to boost healthy growth of neonates. Crit Rev Microbiol 2019; 45:118-129. [PMID: 30773108 DOI: 10.1080/1040841x.2018.1561643] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
For decades, supporting the optimal growth of low birth weight (LBW) infants has been considered one of the most important paediatric challenges, despite advances in neonatal intensive care technology and nutrition interventions. Since gut microbiota affects such diverse phenotypes in adults, the difference in gut microbiota composition between normal infants and LBW infants raises the possibility of gut microbiota playing an important role in different growth rates of neonates. Based on the concept that probiotics are generally beneficial to the health, numerous studies have been made on probiotics as a supplement to the diet of the LBW infants. However, clinical results on the effects of probiotics on LBW infant growth are either inconsistent or contradictory with each other, and thus the contribution of gut microbiota in neonatal growth has remained inconclusive. In this review, recent researches on neonatal gut microbiota are discussed to develop a new strategy for targeting gut microbiota as a solution to growth retardation in LBW infants. We also discuss how to establish the ideal gut microbiota to support optimal growth of LBW infants.
Collapse
Affiliation(s)
- Thi Thanh Binh Nguyen
- a Department of Biomedical Sciences and Institute for Medical Science , Chonbuk National University Medical School , Jeonju , Jeollabuk-do , South Korea.,b Department of Pediatrics , Hue University of Medicine and Pharmacy , Hue , Vietnam
| | - Hea-Jong Chung
- a Department of Biomedical Sciences and Institute for Medical Science , Chonbuk National University Medical School , Jeonju , Jeollabuk-do , South Korea
| | - Hyeon-Jin Kim
- c JINIS BDRD Institute, JINIS Biopharmaceuticals Co , Wanju , Chonbuk , South Korea
| | - Seong-Tshool Hong
- a Department of Biomedical Sciences and Institute for Medical Science , Chonbuk National University Medical School , Jeonju , Jeollabuk-do , South Korea
| |
Collapse
|
21
|
Li X, Peng Y, Li Z, Christensen B, Heckmann AB, Stenlund H, Lönnerdal B, Hernell O. Feeding Infants Formula With Probiotics or Milk Fat Globule Membrane: A Double-Blind, Randomized Controlled Trial. Front Pediatr 2019; 7:347. [PMID: 31552203 PMCID: PMC6736587 DOI: 10.3389/fped.2019.00347] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 08/05/2019] [Indexed: 12/16/2022] Open
Abstract
Purpose: To evaluate effects on growth and infection rates of supplementing infant formula with the probiotic Lactobacillus paracasei ssp. paracasei strain F19 (F19) or bovine milk fat globule membrane (MFGM). Methods: In a double-blind, randomized controlled trial, 600 infants were randomized to a formula supplemented with F19 or MFGM, or to standard formula (SF). A breastfed group was recruited as reference (n = 200).The intervention lasted from age 21 ± 7 days until 4 months, and infants were followed until age one year. Results: Both experimental formulas were well tolerated and resulted in high compliance. The few reported adverse events were not likely related to formula, with the highest rates in the SF group, significantly higher than for the F19-supplemented infants (p = 0.046). Weight or length gain did not differ during or after the intervention among the formula-fed groups, with satisfactory growth. During the intervention, overall, the experimental formula groups did not have more episodes of diarrhea, fever, or days with fever than the breastfed infants. However, compared to the breastfed infants, the SF group had more fever episodes (p = 0.021) and days with fever (p = 0.036), but not diarrhea. Compared with the breastfed group, the F19-supplemented infants but not the other two formula groups had more visits/unscheduled hospitalizations (p = 0.015) and borderline more episodes of upper respiratory tract infections (p = 0.048). Conclusions: Both the MFGM- and F19-supplemented formulas were safe and well-tolerated, leading to few adverse effects, similar to the breastfed group and unlike the SF group. During the intervention, the MFGM-supplemented infants did not differ from the breastfed infants in any primary outcome.
Collapse
Affiliation(s)
- Xiaonan Li
- Department of Children Health Care, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yongmei Peng
- Department of Children Health Care, Children's Hospital of Fudan University, Shanghai, China
| | - Zailing Li
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| | | | | | - Hans Stenlund
- Epidemiology and Global Health, Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Bo Lönnerdal
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Olle Hernell
- Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, Sweden
| |
Collapse
|
22
|
Poinsot P, Schwarzer M, Peretti N, Leulier F. The emerging connections between IGF1, the intestinal microbiome, Lactobacillus strains and bone growth. J Mol Endocrinol 2018; 61:T103-T113. [PMID: 29789323 DOI: 10.1530/jme-17-0292] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 03/29/2018] [Indexed: 12/29/2022]
Abstract
In most animal species, postnatal growth is controlled by conserved insulin/insulin-like growth factor (IGF) signaling. In mammals, juvenile growth is characterized by a longitudinal bone growth resulting from the ossification of the growth plate. This ossification is under IGF1 influence through endocrine and paracrine mechanisms. Moreover, the nutritional status has been largely described as an important factor influencing the insulin/insulin-like growth factor signaling. It is now well established that the gut microbiota modulates the nutrient availability of its host. Hence, studies of the interaction between nutritional status, gut microbiota and bone growth have recently emerged. Here, we review recent findings using experimental models about the impact of gut bacteria on the somatotropic axis and its consequence on the bone growth. We also discuss the perspectives of these studies in opening an entire field for clinical interventions.
Collapse
Affiliation(s)
- Pierre Poinsot
- Institut de Génomique Fonctionnelle de Lyon (IGFL), Université de Lyon, Ecole Normale Supérieure de Lyon, CNRS UMR 5242, Université Claude Bernard Lyon 1, Lyon, France
- Univ Lyon, CarMeN Laboratory, Inserm U1060, INRA U1397, Université Claude Bernard Lyon1, INSA Lyon, Charles Merieux Medical School, Oullins, France
| | - Martin Schwarzer
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Nový Hrádek, Czech Republic
| | - Noël Peretti
- Univ Lyon, CarMeN Laboratory, Inserm U1060, INRA U1397, Université Claude Bernard Lyon1, INSA Lyon, Charles Merieux Medical School, Oullins, France
- Departement of Pediatric Nutrition, Hôpital Femme Mère Enfant, Univ Lyon, Hospice Civil de Lyon, Bron, France
| | - François Leulier
- Institut de Génomique Fonctionnelle de Lyon (IGFL), Université de Lyon, Ecole Normale Supérieure de Lyon, CNRS UMR 5242, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
23
|
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: 37] [Impact Index Per Article: 5.3] [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
|
24
|
|
25
|
Bazanella M, Maier TV, Clavel T, Lagkouvardos I, Lucio M, Maldonado-Gòmez MX, Autran C, Walter J, Bode L, Schmitt-Kopplin P, Haller D. Randomized controlled trial on the impact of early-life intervention with bifidobacteria on the healthy infant fecal microbiota and metabolome. Am J Clin Nutr 2017; 106:1274-1286. [PMID: 28877893 DOI: 10.3945/ajcn.117.157529] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 08/08/2017] [Indexed: 11/14/2022] Open
Abstract
Background: Early-life colonization of the intestinal tract is a dynamic process influenced by numerous factors. The impact of probiotic-supplemented infant formula on the composition and function of the infant gut microbiota is not well defined.Objective: We sought to determine the effects of a bifidobacteria-containing formula on the healthy human intestinal microbiome during the first year of life.Design: A double-blind, randomized, placebo-controlled study of newborn infants assigned to a standard whey-based formula containing a total of 107 colony-forming units (CFU)/g of Bifidobacterium bifidum, Bifidobacterium breve, Bifidobacterium longum, B. longum subspecies infantis (intervention), or to a control formula without bifidobacteria (placebo). Breastfed controls were included. Diversity and composition of fecal microbiota were determined by 16S ribosomal RNA gene amplicon sequencing, and metabolite profiles were analyzed by ultrahigh-performance liquid chromatography-mass spectrometry over a period of 2 y.Results: Infants (n = 106) were randomly assigned to either the interventional (n = 48) or placebo (n = 49) group; 9 infants were exclusively breastfed throughout the entire intervention period of 12 mo. Infants exposed to bifidobacteria-supplemented formula showed decreased occurrence of Bacteroides and Blautia spp. associated with changes in lipids and unknown metabolites at month 1. Microbiota and metabolite profiles of intervention and placebo groups converged during the study period, and long-term colonization (24 mo) of the supplemented Bifidobacterium strains was not detected. Significant differences in microbiota and metabolites were detected between infants fed breast milk and those fed formula (P < 0.005) and between infants birthed vaginally and those birthed by cesarean delivery (P < 0.005). No significant differences were observed between infant feeding groups regarding growth, antibiotic uptake, or other health variables (P > 0.05).Conclusion: The supplementation of bifidobacteria to infant diet can modulate the occurrence of specific bacteria and metabolites during early life with no detectable long-term effects. This trial was registered at germanctr.de as DRKS00003660.
Collapse
Affiliation(s)
| | - Tanja V Maier
- Research Unit Analytical Biogeochemistry, Helmholtz Center Munich, Oberschleißheim, Germany
| | | | | | - Marianna Lucio
- Research Unit Analytical Biogeochemistry, Helmholtz Center Munich, Oberschleißheim, Germany
| | | | - Chloe Autran
- Divisions of Neonatology and Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, and Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (MoMICoRE), University of California, San Diego, La Jolla, CA
| | - Jens Walter
- Chair for Nutrition, Microbes and Gastrointestinal Health, University of Alberta, Edmonton, Alberta, Canada; and
| | - Lars Bode
- Divisions of Neonatology and Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, and Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (MoMICoRE), University of California, San Diego, La Jolla, CA
| | - Philippe Schmitt-Kopplin
- Chair of Analytical Food Chemistry, Technical University of Munich, Freising, Germany.,Research Unit Analytical Biogeochemistry, Helmholtz Center Munich, Oberschleißheim, Germany
| | - Dirk Haller
- Chair of Nutrition and Immunology, .,ZIEL - Institute for Food & Health, and
| |
Collapse
|
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
|
Dror T, Dickstein Y, Dubourg G, Paul M. Microbiota manipulation for weight change. Microb Pathog 2017; 106:146-161. [PMID: 26792677 DOI: 10.1016/j.micpath.2016.01.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 01/07/2016] [Accepted: 01/08/2016] [Indexed: 12/21/2022]
Abstract
Manipulation of the intestinal microbiota has been linked to weight changes and obesity. To explore the influence of specific agents that alter the intestinal flora on weight in different patient groups we conducted a meta-analysis of randomized controlled trials (RCTs) reporting on the effects of probiotics, prebiotics, synbiotics, and antibiotics on weight. We searched the Pubmed and Cochrane Library databases for trials on adults, children, and infants evaluating the effects of these substances on weight. Our primary outcome was weight change from baseline. Standardized mean differences (SMDs) with 95% confidence intervals were calculated. We identified and included 13 adult, 17 children, and 23 infant RCTs. Effects were opposite among adults and children, showing weight loss among adults (SMD -0.54 [-0.83, -0.25)) and minor weight gains among children (SMD 0.20 [0.04, 0.36]) and infants (SMD 0.30 [-0.01, 0.62]) taking mainly Lactobacillus probiotic supplements. Heterogeneity was substantial in the adult and infant analyses and could not be explained by intervention or patient characteristics. Azithromycin administration in children with pulmonary disease was associated with weight gain (SMD 0.39 [0.24, 0.54]), without heterogeneity. A high risk of selective reporting and attrition bias was detected across the studies, making it difficult to draw firm conclusions. Overall, our meta-analysis suggests that there may be a role for probiotics in promoting weight loss in adults and weight gain in children, however additional studies are needed. Though we cannot recommend antibiotic administration for weight manipulation, its use provides advantageous weight gain in children with cystic fibrosis and bronchiectasis.
Collapse
Affiliation(s)
- Tal Dror
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Yaakov Dickstein
- Infectious Diseases Institute, Rambam Health Care Campus, Haifa, Israel
| | - Grégory Dubourg
- Pôle des Maladies Infectieuses et Tropicales Clinique et Biologique, Fédération de Bactériologie-Hygiène-Virologie, University, Hospital Centre Timone, Institut Hospitalo-Universitaire (IHU) Méditerranée Infection, Assistance Publique - Hôpitaux de Marseille, Marseille, France; Université Aix-Marseille, Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes (URMITE) UM 63 CNRS 7278 IRD 198 INSERM U1095, Facultés de Médecine et de Pharmacie, Marseille, France
| | - Mical Paul
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel; Infectious Diseases Institute, Rambam Health Care Campus, Haifa, Israel.
| |
Collapse
|
29
|
Metabolic role of lactobacilli in weight modification in humans and animals. Microb Pathog 2017; 106:182-194. [DOI: 10.1016/j.micpath.2016.03.006] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 03/11/2016] [Accepted: 03/22/2016] [Indexed: 02/07/2023]
|
30
|
Abstract
Malnutrition is the result of an inadequate balance between energy intake and energy expenditure that ultimately leads to either obesity or undernutrition. Several factors are associated with the onset and preservation of malnutrition. One of these factors is the gut microbiota, which has been recognized as an important pathophysiologic factor in the development and sustainment of malnutrition. However, to our knowledge, the extent to which the microbiota influences malnutrition has yet to be elucidated. In this review, we summarize the mechanisms via which the gut microbiota may influence energy homeostasis in relation to malnutrition. In addition, we discuss potential therapeutic modalities to ameliorate obesity or undernutrition.
Collapse
Affiliation(s)
- Nicolien C de Clercq
- Department of Internal and Vascular Medicine, Academic Medical Center, Amsterdam, Netherlands;
| | - Albert K Groen
- Department of Internal and Vascular Medicine, Academic Medical Center, Amsterdam, Netherlands
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Johannes A Romijn
- Department of Internal and Vascular Medicine, Academic Medical Center, Amsterdam, Netherlands
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Academic Medical Center, Amsterdam, Netherlands
- Department of Internal Medicine, Diabetes Center, VU University Medical Center, Amsterdam, Netherlands; and
- Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
31
|
Xu L, Wang Y, Wang Y, Fu J, Sun M, Mao Z, Vandenplas Y. A double‐blinded randomized trial on growth and feeding tolerance with Saccharomyces boulardii CNCM I‐745 in formula‐fed preterm infants. JORNAL DE PEDIATRIA (VERSÃO EM PORTUGUÊS) 2016. [DOI: 10.1016/j.jpedp.2016.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
32
|
Xu L, Wang Y, Wang Y, Fu J, Sun M, Mao Z, Vandenplas Y. A double-blinded randomized trial on growth and feeding tolerance with Saccharomyces boulardii CNCM I-745 in formula-fed preterm infants. J Pediatr (Rio J) 2016; 92:296-301. [PMID: 26946967 DOI: 10.1016/j.jped.2015.08.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 08/10/2015] [Accepted: 08/10/2015] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE The use of probiotics is increasingly popular in preterm neonates, as they may prevent necrotizing enterocolitis sepsis and improve growth and feeding tolerance. There is only limited literature on Saccharomyces boulardii CNCM I-745 (S. boulardii) in preterm infants. METHOD A prospective, randomized, case-controlled trial with the probiotic S. boulardii (50mg/kg twice daily) was conducted in newborns with a gestational age of 30-37 weeks and a birth weight between 1500 and 2500g. RESULTS 125 neonates were enrolled; 63 in the treatment and 62 in the control group. Weight gain (16.14±1.96 vs. 10.73±1.77g/kg/day, p<0.05) and formula intake at maximal enteral feeding (128.4±6.7 vs. 112.3±7.2mL/kg/day, p<0.05) were significantly higher in the intervention group. Once enteral feeding was started, the time needed to reach full enteral feeding was significantly shorter in the probiotic group (0.4±0.1 vs. 1.7±0.5 days, p<0.05). There was no significant difference in sepsis. Necrotizing enterocolitis did not occur. No adverse effects related to S. boulardii were observed. CONCLUSION Prophylactic supplementation of S. boulardii at a dose of 50mg/kg twice a day improved weight gain, improved feeding tolerance, and had no adverse effects in preterm infants >30 weeks old.
Collapse
Affiliation(s)
- Lingfen Xu
- Department of Pediatrics, Shengjing Hospital, China Medical University, Shenyang, China
| | - Yun Wang
- Department of Pediatrics, Qingdao Women and Children's Hospital, Qingdao, China
| | - Yang Wang
- Department of Pediatrics, Shengjing Hospital, China Medical University, Shenyang, China
| | - Jianhua Fu
- Department of Pediatrics, Shengjing Hospital, China Medical University, Shenyang, China
| | - Mei Sun
- Department of Pediatrics, Shengjing Hospital, China Medical University, Shenyang, China
| | - Zhiqin Mao
- Department of Pediatrics, Shengjing Hospital, China Medical University, Shenyang, China.
| | - Yvan Vandenplas
- UZ Brussel, Department of Pediatrics, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
33
|
López-Huertas E. Safety and efficacy of human breast milk Lactobacillus fermentum CECT 5716. A mini-review of studies with infant formulae. Benef Microbes 2016; 6:219-24. [PMID: 25519525 DOI: 10.3920/bm2014.0091] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Human breast milk has been described as a source of lactic acid bacteria. Lactobacillus fermentum CECT5716 is a human breast milk strain whose probiotic properties, safety and efficacy has been demonstrated in vitro and in vivo, including controlled trials with human adults. Since the origin of this probiotic strain is human breast milk, we aimed to investigate the safety and efficacy of an infant and a follow-on formulas supplemented with this strain of L. fermentum. We carried out two randomised controlled trials: one trial with infants of 6-12 months of age (follow-on formula study) and another one with infants from 1 to 5 months of age (infant formula study). The results from the trials showed that the probiotic formulas were safe, well tolerated and might be useful for the prevention of community-acquired infections.
Collapse
Affiliation(s)
- E López-Huertas
- Estación Experimental Zaidín, Spanish Council for Scientific Research, Profesor Albareda 1, 18008 Granada, Spain
| |
Collapse
|
34
|
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
|
35
|
Contopoulos-Ioannidis DG, Ley C, Wang W, Ma T, Olson C, Shi X, Luft HS, Hastie T, Parsonnet J. Effect of long-term antibiotic use on weight in adolescents with acne. J Antimicrob Chemother 2016; 71:1098-105. [PMID: 26782773 DOI: 10.1093/jac/dkv455] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 11/29/2015] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVES Antibiotics increase weight in farm animals and may cause weight gain in humans. We used electronic health records from a large primary care organization to determine the effect of antibiotics on weight and BMI in healthy adolescents with acne. METHODS We performed a retrospective cohort study of adolescents with acne prescribed ≥4 weeks of oral antibiotics with weight measurements within 18 months pre-antibiotics and 12 months post-antibiotics. We compared within-individual changes in weight-for-age Z-scores (WAZs) and BMI-for-age Z-scores (BMIZs). We used: (i) paired t-tests to analyse changes between the last pre-antibiotics versus the first post-antibiotic measurements; (ii) piecewise-constant-mixed models to capture changes between mean measurements pre- versus post-antibiotics; (iii) piecewise-linear-mixed models to capture changes in trajectory slopes pre- versus post-antibiotics; and (iv) χ(2) tests to compare proportions of adolescents with ≥0.2 Z-scores WAZ or BMIZ increase or decrease. RESULTS Our cohort included 1012 adolescents with WAZs; 542 also had BMIZs. WAZs decreased post-antibiotics in all analyses [change between last WAZ pre-antibiotics versus first WAZ post-antibiotics = -0.041 Z-scores (P < 0.001); change between mean WAZ pre- versus post-antibiotics = -0.050 Z-scores (P < 0.001); change in WAZ trajectory slopes pre- versus post-antibiotics = -0.025 Z-scores/6 months (P = 0.002)]. More adolescents had a WAZ decrease post-antibiotics ≥0.2 Z-scores than an increase (26% versus 18%; P < 0.001). Trends were similar, though not statistically significant, for BMIZ changes. CONCLUSIONS Contrary to original expectations, long-term antibiotic use in healthy adolescents with acne was not associated with weight gain. This finding, which was consistent across all analyses, does not support a weight-promoting effect of antibiotics in adolescents.
Collapse
Affiliation(s)
- Despina G Contopoulos-Ioannidis
- Division of Infectious Diseases, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA Palo Alto Medical Foundation Research Institute, Palo Alto, CA 94301, USA
| | - Catherine Ley
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Wei Wang
- Palo Alto Medical Foundation Research Institute, Palo Alto, CA 94301, USA
| | - Ting Ma
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Clifford Olson
- Palo Alto Medical Foundation Research Institute, Palo Alto, CA 94301, USA
| | - Xiaoli Shi
- Palo Alto Medical Foundation Research Institute, Palo Alto, CA 94301, USA
| | - Harold S Luft
- Palo Alto Medical Foundation Research Institute, Palo Alto, CA 94301, USA
| | - Trevor Hastie
- Department of Statistics, Stanford University School of Humanities and Sciences and Department of Health Research and Policy, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Julie Parsonnet
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA Department of Health Research and Policy, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
36
|
Balasubramanian H, Patole S. Early probiotics to prevent childhood metabolic syndrome: A systematic review. World J Methodol 2015; 5:157-163. [PMID: 26413489 PMCID: PMC4572029 DOI: 10.5662/wjm.v5.i3.157] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 04/05/2015] [Accepted: 06/19/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To conduct a systematic review of studies on early probiotic supplementation to prevent childhood metabolic syndrome (MS).
METHODS: Using the Cochrane systematic review strategy we searched PubMed, EMBASE, CENTRAL, CINAHL, and the conference proceedings of the Pediatric American Society meetings and trial registries in December 2014. Randomised controlled trials (RCTs) and non RCTs of probiotic supplementation to the mother and/or infant for a minimum duration of 4 wk were selected. Of these, studies that reported on MS or its components (obesity, raised blood pressure, hyperglycemia, dyslipidemia) in children between 2-19 years were to be eligible for inclusion in the review. Risk of bias (ROB) in selected RCTs and quality assessment of non-RCT studies were to be assessed by the Cochrane ROB assessment table and New Castle Ottawa scale.
RESULTS: There were no studies on early probiotic administration for prevention of childhood MS (CMS). Follow up studies of two placebo controlled RCTs (n = 233) reported on the effects of early probiotics on one or more components of MS in children aged 2-19 years. Meta-analysis of those two studies could not be performed due to differences in the patient population, type of outcomes studied and the timing of their assessment. Assessment of childhood metabolic outcomes was not the primary objective of these studies. The first study that assessed the effects of prenatal and postnatal supplementation of Lactobacillus rhamnosus GG on body mass index till 10 years, did not report a significant benefit. In the second study, Lactobacillus paracasei 19 was supplemented to healthy term infants from 4-13 mo. No significant effect on body mass index, body composition or metabolic markers was detected.
CONCLUSION: Current evidence on early probiotic administration to prevent CMS is inadequate. Gaps in knowledge need to be addressed before large RCTs can be planned.
Collapse
|
37
|
Aw W, Fukuda S. The Role of Integrated Omics in Elucidating the Gut Microbiota Health Potentials. ACTA ACUST UNITED AC 2015. [DOI: 10.1007/978-3-319-23213-3_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
38
|
Aw W, Fukuda S. Toward the comprehensive understanding of the gut ecosystem via metabolomics-based integrated omics approach. Semin Immunopathol 2014; 37:5-16. [PMID: 25338280 DOI: 10.1007/s00281-014-0456-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 10/09/2014] [Indexed: 12/27/2022]
Abstract
Recent advances in DNA sequencing and mass spectrometry technologies have allowed us to collect more data on microbiome and metabolome to assess the influence of the gut microbiota on human health at a whole-systems level. Major advances in metagenomics and metabolomics technologies have shown that the gut microbiota contributes to host overall health status to a large extent. As such, the gut microbiota is often likened to a measurable and functional organ consisting of prokaryotic cells, which creates the unique gut ecosystem together with the host eukaryotic cells. In this review, we discuss in detail the relationship between gut microbiota and its metabolites like choline, bile acids, phenols, and short-chain fatty acids in the host health and etiopathogenesis of various pathological states such as multiple sclerosis, autism, obesity, diabetes, and chronic kidney disease. By integrating metagenomic and metabolomic information on a systems biology-wide approach, we would be better able to understand this interplay between gut microbiome and host metabolism. Integration of the microbiome, metatranscriptome, and metabolome information will pave the way toward an improved holistic understanding of the complex mammalian superorganism. Through the modeling of metabolic interactions between lifestyle, diet, and microbiota, integrated omics-based understanding of the gut ecosystem is the new avenue, providing exciting novel therapeutic approaches for optimal host health.
Collapse
Affiliation(s)
- Wanping Aw
- Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka, Yamagata, 997-0052, Japan
| | | |
Collapse
|
39
|
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]
|
40
|
Druart C, Alligier M, Salazar N, Neyrinck AM, Delzenne NM. Modulation of the gut microbiota by nutrients with prebiotic and probiotic properties. Adv Nutr 2014; 5:624S-633S. [PMID: 25225347 PMCID: PMC4188246 DOI: 10.3945/an.114.005835] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Experimental data in animals, but also observational studies in humans, suggest that the composition of the gut microbiota differs in obese vs. lean individuals, in patients with vs. without diabetes, or in patients presenting other diseases associated with obesity or nutritional disbalance, such as non-alcoholic fatty liver disease (NAFLD) or cardiovascular diseases. In this review, we describe how changes in the composition and/or activity of the gut microbiota by administration of nutrients with probiotic or prebiotic properties can modulate host gene expression and metabolism and thereby positively influence host adipose tissue development and related metabolic disorders.
Collapse
|
41
|
Kumar A, Vlasova AN, Liu Z, Chattha KS, Kandasamy S, Esseili M, Zhang X, Rajashekara G, Saif LJ. In vivo gut transcriptome responses to Lactobacillus rhamnosus GG and Lactobacillus acidophilus in neonatal gnotobiotic piglets. Gut Microbes 2014; 5:152-64. [PMID: 24637605 PMCID: PMC4063840 DOI: 10.4161/gmic.27877] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Probiotics facilitate mucosal repair and maintain gut homeostasis. They are often used in adjunct with rehydration or antibiotic therapy in enteric infections. Lactobacillus spp have been tested in infants for the prevention or treatment of various enteric conditions. However, to aid in rational strain selection for specific treatments, comprehensive studies are required to delineate and compare the specific molecules and pathways involved in a less complex but biologically relevant model (gnotobiotic pigs). Here we elucidated Lactobacillus rhamnosus (LGG) and L. acidophilus (LA) specific effects on gut transcriptome responses in a neonatal gnotobiotic (Gn) pig model to simulate responses in newly colonized infants. Whole genome microarray, followed by biological pathway reconstruction, was used to investigate the host-microbe interactions in duodenum and ileum at early (day 1) and later stages (day 7) of colonization. Both LA and LGG modulated common responses related to host metabolism, gut integrity, and immunity, as well as responses unique to each strain in Gn pigs. Our data indicated that probiotic establishment and beneficial effects in the host are guided by: (1) down-regulation or upregulation of immune function-related genes in the early and later stages of colonization, respectively, and (2) alternations in metabolism of small molecules (vitamins and/or minerals) and macromolecules (carbohydrates, proteins, and lipids). Pathways related to immune modulation and carbohydrate metabolism were more affected by LGG, whereas energy and lipid metabolism-related transcriptome responses were prominently modulated by LA. These findings imply that identification of probiotic strain-specific gut responses could facilitate the rational design of probiotic-based interventions to moderate specific enteric conditions.
Collapse
Affiliation(s)
- Anand Kumar
- Food Animal Health Research Program; Department of Veterinary Preventive Medicine; Ohio Agricultural Research and Development Center; The Ohio State University; Wooster, OH USA
| | - Anastasia N Vlasova
- Food Animal Health Research Program; Department of Veterinary Preventive Medicine; Ohio Agricultural Research and Development Center; The Ohio State University; Wooster, OH USA
| | - Zhe Liu
- Food Animal Health Research Program; Department of Veterinary Preventive Medicine; Ohio Agricultural Research and Development Center; The Ohio State University; Wooster, OH USA
| | - Kuldeep S Chattha
- Food Animal Health Research Program; Department of Veterinary Preventive Medicine; Ohio Agricultural Research and Development Center; The Ohio State University; Wooster, OH USA
| | - Sukumar Kandasamy
- Food Animal Health Research Program; Department of Veterinary Preventive Medicine; Ohio Agricultural Research and Development Center; The Ohio State University; Wooster, OH USA
| | - Malak Esseili
- Food Animal Health Research Program; Department of Veterinary Preventive Medicine; Ohio Agricultural Research and Development Center; The Ohio State University; Wooster, OH USA
| | - Xiaoli Zhang
- Center for Biostatistics; The Ohio State University; Columbus, OH USA
| | - Gireesh Rajashekara
- Food Animal Health Research Program; Department of Veterinary Preventive Medicine; Ohio Agricultural Research and Development Center; The Ohio State University; Wooster, OH USA,Correspondence to: Gireesh Rajashekara, and Linda J Saif,
| | - Linda J Saif
- Food Animal Health Research Program; Department of Veterinary Preventive Medicine; Ohio Agricultural Research and Development Center; The Ohio State University; Wooster, OH USA,Correspondence to: Gireesh Rajashekara, and Linda J Saif,
| |
Collapse
|
42
|
Tijhuis MJ, Doets EL, Vonk Noordegraaf‐Schouten M. Extensive literature search and review as preparatory work for the evaluation of the essential composition of infant and follow‐on formulae and growing‐up milk. ACTA ACUST UNITED AC 2014. [DOI: 10.2903/sp.efsa.2014.en-551] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- MJ Tijhuis
- Pallas health research and consultancy the Netherlands
| | - EL Doets
- Pallas health research and consultancy the Netherlands
| | | |
Collapse
|
43
|
Angelakis E, Merhej V, Raoult D. Related actions of probiotics and antibiotics on gut microbiota and weight modification. THE LANCET. INFECTIOUS DISEASES 2013; 13:889-99. [PMID: 24070562 DOI: 10.1016/s1473-3099(13)70179-8] [Citation(s) in RCA: 136] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Antibiotics and probiotics are widely used as growth promoters in agriculture. Most antibiotics prescribed in clinical practice are natural products that originate from Streptomyces spp, which were first used as agricultural probiotics. Antibiotics and probiotics both modify the gut microbiota. The effect of a probiotic species on the digestive flora depends on the strain and is largely determined by bacteriocin production. In human beings, as in animals, specific probiotics are associated with weight gain or loss. Improved understanding of the ability of specific probiotics to harvest energy from the host diet might lead to development of new treatments for obesity and malnutrition. In this Review, we present the effects of probiotics and antibiotics on the gut microbiota of human beings and animals and discuss their potential therapeutic use as interventions for weight gain and loss in human beings.
Collapse
Affiliation(s)
- Emmanouil Angelakis
- Unité des Rickettsies, Faculté de Médecine, Université de la Méditerranée, Marseille, France
| | | | | |
Collapse
|
44
|
Mugambi MN, Musekiwa A, Lombard M, Young T, Blaauw R. Association between funding source, methodological quality and research outcomes in randomized controlled trials of synbiotics, probiotics and prebiotics added to infant formula: a systematic review. BMC Med Res Methodol 2013; 13:137. [PMID: 24219082 PMCID: PMC3832685 DOI: 10.1186/1471-2288-13-137] [Citation(s) in RCA: 18] [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: 07/19/2013] [Accepted: 11/07/2013] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND There is little or no information available on the impact of funding by the food industry on trial outcomes and methodological quality of synbiotics, probiotics and prebiotics research in infants. The objective of this study was to compare the methodological quality, outcomes of food industry sponsored trials versus non industry sponsored trials, with regards to supplementation of synbiotics, probiotics and prebiotics in infant formula. METHODS A comprehensive search was conducted to identify published and unpublished randomized clinical trials (RCTs). Cochrane methodology was used to assess the risk of bias of included RCTs in the following domains: 1) sequence generation; 2) allocation concealment; 3) blinding; 4) incomplete outcome data; 5) selective outcome reporting; and 6) other bias. Clinical outcomes and authors' conclusions were reported in frequencies and percentages. The association between source of funding, risk of bias, clinical outcomes and conclusions were assessed using Pearson's Chi-square test and the Fisher's exact test. A p-value < 0.05 was statistically significant. RESULTS Sixty seven completed and 3 on-going RCTs were included. Forty (59.7%) were funded by food industry, 11 (16.4%) by non-industry entities and 16 (23.9%) did not specify source of funding. Several risk of bias domains, especially sequence generation, allocation concealment and blinding, were not adequately reported. There was no significant association between the source of funding and sequence generation, allocation concealment, blinding and selective reporting, majority of reported clinical outcomes or authors' conclusions. On the other hand, source of funding was significantly associated with the domains of incomplete outcome data, free of other bias domains as well as reported antibiotic use and conclusions on weight gain. CONCLUSION In RCTs on infants fed infant formula containing probiotics, prebiotics or synbiotics, the source of funding did not influence the majority of outcomes in favour of the sponsors' products. More non-industry funded research is needed to further assess the impact of funding on methodological quality, reported clinical outcomes and authors' conclusions.
Collapse
Affiliation(s)
- Mary N Mugambi
- Division of Human Nutrition, Faculty of Medicine and Health Sciences, Stellenbosch University, P.O Box 19063, Tygerberg 7505, South Africa
| | - Alfred Musekiwa
- Centre for Evidence-Based Health Care, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Martani Lombard
- Division of Human Nutrition, Faculty of Medicine and Health Sciences, Stellenbosch University, P.O Box 19063, Tygerberg 7505, South Africa
| | - Taryn Young
- Centre for Evidence-Based Health Care, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Reneé Blaauw
- Division of Human Nutrition, Faculty of Medicine and Health Sciences, Stellenbosch University, P.O Box 19063, Tygerberg 7505, South Africa
| |
Collapse
|
45
|
Szajewska H, Chmielewska A. Growth of infants fed formula supplemented with Bifidobacterium lactis Bb12 or Lactobacillus GG: a systematic review of randomized controlled trials. BMC Pediatr 2013; 13:185. [PMID: 24215626 PMCID: PMC3831250 DOI: 10.1186/1471-2431-13-185] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 10/31/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Growth is an essential outcome measure for evaluating the safety of any new ingredients, including probiotics, added to infant formulae. The aim of this systematic review was to determine the effects of supplementation of infant formulae with Bifidobacterium lactis Bb12 (B lactis) and/or Lactobacillus rhamnosus GG (LGG) compared with unsupplemented formula on the growth of healthy infants. METHODS The MEDLINE, EMBASE, and Cochrane Library databases were searched in June 2013 for relevant randomized controlled trials (RCTs) conducted in healthy term infants. Unpublished data were obtained from the manufacturer of B lactis-supplemented formula. The primary outcome measures were weight, length, and head circumference. RESULTS Nine eligible trials were identified. Compared with unsupplemented controls, supplementation of infant formula with B lactis had no effect on weight gain [4 RCTs, n = 266, mean difference (MD) 0.96 g/day, 95% confidence interval (CI) -0.70 to 2.63)], length gain (4 RCTs, n = 261, MD -0.39 mm/month, 95% CI -1.32 to 0.53), or head circumference gain (3 RCTs, n = 207, MD 0.56 mm/month, 95% CI -0.17 to 1.30). Data limited to one small (n = 105) trial suggest that infants who received standard infant formula supplemented with LGG grew significantly better. No such effect was observed in infants fed hydrolyzed formula supplemented with LGG. CONCLUSIONS Supplementation of infant formula with B lactis results in growth similar to what is found in infants fed unsupplemented formula. Limited data do not allow one to reach a conclusion regarding the effect of LGG supplementation on infant growth.
Collapse
Affiliation(s)
- Hania Szajewska
- Department of Paediatrics, The Medical University of Warsaw, Dzialdowska 1, Warsaw 01-183, Poland.
| | | |
Collapse
|
46
|
Fukuda S, Ohno H. Gut microbiome and metabolic diseases. Semin Immunopathol 2013; 36:103-14. [PMID: 24196453 DOI: 10.1007/s00281-013-0399-z] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Accepted: 10/13/2013] [Indexed: 02/06/2023]
Abstract
The prevalence of obesity and obesity-related disorders is increasing worldwide. In the last decade, the gut microbiota has emerged as an important factor in the development of obesity and metabolic syndrome, through its interactions with dietary, environmental, and host genetic factors. Various studies have shown that alteration of the gut microbiota, shifting it toward increased energy harvest, is associated with an obese phenotype. However, the molecular mechanisms by which the gut microbiota affects host metabolism are still obscure. In this review, we discuss the complexity of the gut microbiota and its relationship to obesity and obesity-related diseases. Furthermore, we discuss the anti-obesity potential of probiotics and prebiotics.
Collapse
Affiliation(s)
- Shinji Fukuda
- Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka, Yamagata, 997-0052, Japan
| | | |
Collapse
|
47
|
Pyar H, Liong MT, Peh K. Recent Advances in Probiotics and Biomedical Applications. JOURNAL OF MEDICAL SCIENCES 2013. [DOI: 10.3923/jms.2013.601.614] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
48
|
Civardi E, Garofoli F, Tzialla C, Paolillo P, Bollani L, Stronati M. Microorganisms in human milk: lights and shadows. J Matern Fetal Neonatal Med 2013; 26 Suppl 2:30-4. [DOI: 10.3109/14767058.2013.829693] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
49
|
Bernardeau M, Vernoux JP. Overview of differences between microbial feed additives and probiotics for food regarding regulation, growth promotion effects and health properties and consequences for extrapolation of farm animal results to humans. Clin Microbiol Infect 2013; 19:321-30. [DOI: 10.1111/1469-0691.12130] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Revised: 12/11/2012] [Accepted: 12/12/2012] [Indexed: 12/28/2022]
|
50
|
Ostadrahimi A, Nikniaz L, Mahdavi R, Hejazi MA, Nikniaz Z. Effects of synbiotic supplementation on lactating mothers' energy intake and BMI, and infants' growth. Int J Food Sci Nutr 2013; 64:711-4. [PMID: 23480276 DOI: 10.3109/09637486.2013.775229] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In this randomized, double-blind, placebo-controlled trial, 80 lactating mothers were randomly divided into two groups to receive daily supplement of synbiotic (n = 40) or a placebo (n = 40) for 30 days. Information on dietary intake was collected and anthropometric measurements were taken using standard calibrated instruments. Data analysis was carried out using nutritionist IV, SPSS and Epi Info software. Synbiotic supplementation resulted in a slight increase in mean energy intake while, in the placebo group, maternal energy intake decreased significantly (p < 0.023). Although maternal weight and BMI increased slightly in the supplemented group, these two parameters decreased significantly in the placebo group (p < 0.01). Also, infants' weight gain in the synbiotic group was significantly higher than the placebo group after the intervention (p < 0.044). Synbiotics may prevent weight loss in lactating mothers and result in weight gain in infants. Further experiments are required to study these effects in undernourished lactating mothers and their infants.
Collapse
Affiliation(s)
- Alireza Ostadrahimi
- Nutrition Research Center, School of Public Health & Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | | | | |
Collapse
|