1
|
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
|
2
|
Amar Y, Grube J, Köberle M, Schaubeck M, Biedermann T, Volz T. Bifidobacterium breve DSM 32583 and Limosilactobacillus fermentum CECT5716 postbiotics attenuate S. aureus and IL-33-induced Th2 responses. Microbiol Res 2024; 289:127913. [PMID: 39316930 DOI: 10.1016/j.micres.2024.127913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/04/2024] [Accepted: 09/15/2024] [Indexed: 09/26/2024]
Abstract
Over the past decades, the prevalence of allergic diseases noticeably increased in industrialized countries. The Th2 immune response plays a central role in these pathologies and its modulation using pro-/postbiotics constitutes a promising approach to prevent or alleviate disease symptoms. The aim of this in vitro study, was to investigate the ability of human milk-derived Bifidobacterium breve DSM 32583 (Bb) and Limosilactobacillus fermentum CECT5716 (Lf), to modulate the Th2 induced responses. To this end, Th2 cells were generated by co-culturing of human naïve Th cells with monocyte-derived dendritic cells (moDCs) either stimulated with Staphylococcus aureus or IL-33. The immunomodulatory effects of pro-/postbiotic preparations of Bb and Lf on moDCs and Th2 cells were evaluated in terms of maturation markers expression and cytokines production. Remarkably, the tested strains induced the anti-inflammatory cytokine IL-10 in moDCs, in a strain-, dose- and viability-dependent manner with no significant upregulation of IL-12p70 nor CD83, CD86 or HLA-DR. Interestingly, Bb and Lf postbiotics were able to dampen the Th2/Th1 response induced upon S. aureus- or IL-33 stimulation. They were also able to synergistically induce IL-10 in moDCs and T cells, upon co-stimulation with LPS. Finally, we observed that live probiotics triggered a mild Th1 response that was attenuated in the presence of galacto-oligosaccharides. Altogether, Bb and Lf pro-/postbiotics exhibited remarkable immune regulatory effects on both moDCs and Th2 cells. Therefore, further in vivo studies should be considered to validate these findings and assess their ability to prevent allergy or alleviate its symptoms in affected patients.
Collapse
Affiliation(s)
- Yacine Amar
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich 80802, Germany.
| | - Jana Grube
- HiPP GmbH & Co. Vertrieb KG, Pfaffenhofen (Ilm) 85276, Germany
| | - Martin Köberle
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich 80802, Germany
| | | | - Tilo Biedermann
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich 80802, Germany
| | - Thomas Volz
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich 80802, Germany
| |
Collapse
|
3
|
Zhang W, Zhang Y, Zhao Y, Li L, Zhang Z, Hettinga K, Yang H, Deng J. A Comprehensive Review on Dietary Polysaccharides as Prebiotics, Synbiotics, and Postbiotics in Infant Formula and Their Influences on Gut Microbiota. Nutrients 2024; 16:4122. [PMID: 39683515 DOI: 10.3390/nu16234122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Human milk contains an abundance of nutrients which benefit the development and growth of infants. However, infant formula has to be used when breastfeeding is not possible. The large differences between human milk and infant formula in prebiotics lead to the suboptimal intestinal health of infant formula-fed infants. This functional deficit of infant formula may be overcome through other dietary polysaccharides that have been characterized. The aim of this review was to summarize the potential applications of dietary polysaccharides as prebiotics, synbiotics, and postbiotics in infant formula to better mimic the functionality of human milk prebiotics for infant gut health. Previous studies have demonstrated the influences of dietary polysaccharides on gut microbiota, SCFA production, and immune system development. Compared to prebiotics, synbiotics and postbiotics showed better application potential in shaping the gut microbiota, the prevention of pathogen infections, and the development of the immune system. Moreover, the safety issues for biotics still require more clinical trials with a large-scale population and long time duration, and the generally accepted regulations are important to regulate related products. Pectin polysaccharides has similar impacts to human milk oligosaccharides on gut microbiota and the repairing of a damaged gut barrier, with similar functions also being observed for inulin and β-glucan. Prebiotics as an encapsulation material combined with probiotics and postbiotics showed better potential applications compared to traditional material in infant formula.
Collapse
Affiliation(s)
- Wenyuan Zhang
- State Key Laboratory of Vegetable Biobreeding, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yanli Zhang
- State Key Laboratory of Vegetable Biobreeding, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yaqi Zhao
- State Key Laboratory of Vegetable Biobreeding, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Liang Li
- State Key Laboratory of Vegetable Biobreeding, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Zhanquan Zhang
- State Key Laboratory of Vegetable Biobreeding, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Kasper Hettinga
- Dairy Science and Technology, Food Quality and Design Group, Wageningen University & Research, 6708 WG Wageningen, The Netherlands
| | - Haixia Yang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Jianjun Deng
- State Key Laboratory of Vegetable Biobreeding, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| |
Collapse
|
4
|
Yang R, Shi Z, Li Y, Huang X, Li Y, Li X, Chen Q, Hu Y, Li X. Research focus and emerging trends of the gut microbiome and infant: a bibliometric analysis from 2004 to 2024. Front Microbiol 2024; 15:1459867. [PMID: 39633813 PMCID: PMC11615055 DOI: 10.3389/fmicb.2024.1459867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024] Open
Abstract
Background Over the past two decades, gut microbiota has demonstrated unprecedented potential in human diseases and health. The gut microbiota in early life is crucial for later health outcomes. This study aims to reveal the knowledge collaboration network, research hotspots, and explore the emerging trends in the fields of infant and gut microbiome using bibliometric analysis. Method We searched the literature on infant and gut microbiome in the Web of Science Core Collection (WOSCC) database from 2004 to 2024. CiteSpace V (version: 6.3.R1) and VOSview (version: 1.6.20) were used to display the top authors, journals, institutions, countries, authors, keywords, co-cited articles, and potential trends. Results A total of 9,899 documents were retrieved from the Web of Science Core Collection. The United States, China, and Italy were the three most productive countries with 3,163, 1,510, and 660 publications. The University of California System was the most prolific institution (524 publications). Van Sinderen, Douwe from University College Cork of Ireland was the most impactful author. Many studies have focused on atopic dermatitis (AD), necrotizing enterocolitis (NEC), as well as the immune mechanisms and microbial treatments for these diseases, such as probiotic strains mixtures and human milk oligosaccharides (HMOs). The mother-to-infant microbiome transmission, chain fatty acids, and butyrate maybe the emerging trends. Conclusion This study provided an overview of the knowledge structure of infant and gut microbiome, as well as a reference for future research.
Collapse
Affiliation(s)
- Ru Yang
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Zeyao Shi
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Yuan Li
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Xi Huang
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Yingxin Li
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Xia Li
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Qiong Chen
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Yanling Hu
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Xiaowen Li
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| |
Collapse
|
5
|
Anuk Ince D, Takci S, Kilicdag H, Turan O. Editorial: Feeding difficulties in newborn infants and new approaches in practice. Front Pediatr 2024; 12:1462493. [PMID: 39301038 PMCID: PMC11410623 DOI: 10.3389/fped.2024.1462493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 08/28/2024] [Indexed: 09/22/2024] Open
Affiliation(s)
- Deniz Anuk Ince
- Division of Neonatology, Department of Pediatrics, Baskent University Faculty of Medicine, Ankara, Türkiye
| | - Sahin Takci
- Division of Neonatology, Department of Pediatrics, Ondokuz Mayıs University Faculty of Medicine, Samsun, Türkiye
| | - Hasan Kilicdag
- Division of Neonatology, Department of Pediatrics, Baskent University Faculty of Medicine, Ankara, Türkiye
| | - Ozden Turan
- Division of Neonatology, Department of Pediatrics, Baskent University Faculty of Medicine, Ankara, Türkiye
| |
Collapse
|
6
|
Bognanni A, Firmino RT, Arasi S, Chu DK, Chu AW, Waffenschmidt S, Agarwal A, Dziechciarz P, Horvath A, Mihara H, Roldan Y, Terracciano L, Martelli A, Starok A, Said M, Shamir R, Ansotegui IJ, Dahdah L, Ebisawa M, Galli E, Kamenwa R, Lack G, Li H, Pawankar R, Warner A, Wong GWK, Bozzola M, Assa'Ad A, Dupont C, Bahna S, Spergel J, Venter C, Szajewska H, Nowak-Wegrzyn AH, Vandenplas Y, Papadopoulos NG, Waserman S, Fiocchi A, Schünemann HJ, Brożek JL. World Allergy Organization (WAO) Diagnosis and Rationale for Action against Cow's Milk Allergy (DRACMA) guideline update - XI - Milk supplement/replacement formulas for infants and toddlers with CMA - Systematic review. World Allergy Organ J 2024; 17:100947. [PMID: 39310372 PMCID: PMC11415968 DOI: 10.1016/j.waojou.2024.100947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 09/25/2024] Open
Abstract
Background Cow's milk allergy (CMA) is the most complex and common food allergy in infants. Elimination of cow's milk from the diet and replacement with a specialized formula for infants with cow's milk allergy who cannot be breastfed is an established approach to minimize the risk of severe allergic reactions while avoiding nutritional deficiencies. Given the availability of multiple options, such as extensively hydrolyzed cow's milk-based formula (eHF-CM), aminoacid formula (AAF), hydrolyzed rice formula (HRF), and soy formula (SF), there is some uncertainty regarding which formula might represent the most suitable choice with respect to health outcomes. The addition of probiotics to a specialized formula has also been proposed as a potential approach to possibly increase the benefit. We systematically reviewed specialized formulas for infants with CMA to inform the updated World Allergy Organization (WAO) DRACMA guidelines. Objective To systematically review and synthesize the available evidence about the use of specialized formulas for the management of individuals with CMA. Methods We searched from inception PubMed, Medline, Embase, the Cochrane Central Register of Controlled Trials (CENTRAL), and the websites of selected allergy organizations, for randomized and non-randomized trials of any language investigating specialized formulas with or without probiotics. We included all studies irrespective of the language of the original publication. The last search was conducted in January 2024. We synthesized the identified evidence quantitatively or narratively as appropriate and summarized it in the evidence profiles. We conducted this review following the PRISMA, Cochrane methods, and the GRADE approach. Results We identified 3558 records including 14 randomized trials and 7 observational studies. Very low certainty evidence suggested that in infants with IgE-mediated CMA, eHF-CM, compared with AAF, might have higher probability of outgrowing CMA (risk ratio (RR) 2.32; risk difference (RD) 25 more per 100), while showing potentially lower probability of severe vomiting (RR 0.12, 95% CI 0.02 to 0.88; RD 23 fewer per 100, 95% CI 3 to 26) and developing food protein-induced enterocolitis syndrome (FPIES) (RR 0.15, 95% CI 0.03 to 0.82; RD 34 fewer per 100, 95% CI 7 to 39). We also found, however, that eHF-CM might be inferior to AAF in supporting a physiological growth, with respect to both weight (-5.5% from baseline, 95%CI -9.5% to -1.5%) and length (-0.7 z-score change, 95%CI -1.15 to -0.25) (very low certainty). We found similar effects for eHF-CM, compared with AAF, also in non-IgE CMA. When compared with SF, eHF-CM might favor weight gain for IgE CMA infants (0.23 z-score change, 95%CI 0.01 to 0.45), and tolerance acquisition (RR 1.86, 95%CI 1.03 to 3.37; RD 27%, 95%CI 1%-74%) for non-IgE CMA (both at very low certainty of the evidence (CoE)). The comparison of eHF-CM vs. HRF, and HRF vs. SF, showed no difference in effect (very low certainty). For IgE CMA patients, low certainty evidence suggested that adding probiotics (L. rhamnosus GG, L. casei CRL431 and B. lactis Bb-12) might increase the probability of developing CMA tolerance (RR 2.47, 95%CI 1.03 to 5.93; RD 27%, 95%CI 1%-91%), and reduce the risk of severe wheezing (RR 0.12, 95%CI 0.02 to 0.95; RD -23%, 95%CI -8% to -0.4%). However, in non-IgE CMA infants, the addition of probiotics (L. rhamnosus GG) showed no significant effect, as supported by low to very low CoE. Conclusions Currently available studies comparing eHF-CM, AAF, HRF, and SF provide very low certainty evidence about their effects in infants with IgE-mediated and non-IgE-mediated CMA. Our review revealed several limitations in the current body of evidence, primarily arising from concerns related to the quality of studies, the limited size of the participant populations and most importantly the lack of diversity and standardization in the compared interventions. It is therefore imperative for future studies to be methodologically rigorous and investigate a broader spectrum of available interventions. We encourage clinicians and researchers to review current World Allergy Organization (WAO) Diagnosis and Rationale for Action against Cow's Milk Allergy (DRACMA) Guidelines for suggestions on how to use milk replacement formulas in clinical practice and what additional research would be the most beneficial.
Collapse
Affiliation(s)
- Antonio Bognanni
- Department of Health Research Methods, Evidence & Impact, McMaster University, Hamilton, Ontario, Canada
- Clinical Epidemiology and Research Center (CERC), Humanitas University & Humanitas Research Hospital, Pieve Emanuele, Milano, Italy
- Department of Medicine, Evidence in Allergy Group, McMaster University, Hamilton, Ontario, Canada
| | - Ramon T. Firmino
- Academic Unit of Biological Sciences, Federal University of Campina Grande, Patos, Paraíba, Brazil
| | - Stefania Arasi
- Translational Research in Pediatric Specialties Area, Division of Allergy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Derek K. Chu
- Department of Health Research Methods, Evidence & Impact, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, Evidence in Allergy Group, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, Division of Clinical Immunology and Allergy, McMaster University, Hamilton, Ontario, Canada
| | - Alexandro W.L. Chu
- Department of Medicine, Evidence in Allergy Group, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Siw Waffenschmidt
- Institute for Quality and Efficiency in Health Care, Cologne, Germany
| | - Arnav Agarwal
- Department of Medicine, Division of Internal Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | | | - Hanako Mihara
- Clinical Development Infectious Disease, Moderna Japan Co., Ltd., Japan
| | | | - Luigi Terracciano
- Pediatric Primary Care, National Pediatric Health Care System, Milan, Italy
- Italian Society of Preventive and Social Pediatrics (SIPPS), Italy
| | - Alberto Martelli
- Italian Society of Allergy and Pediatric Immunology (SIAIP), Italy
| | | | - Maria Said
- Allergy & Anaphylaxis Australia, Sydney, Australia
| | - Raanan Shamir
- Institute for Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center of Israel, Faculty of Medical and Health Sciences, Tel Aviv University, Israel
| | - Ignacio J. Ansotegui
- Department of Allergy & Immunology, Hospital Quironsalud Bizkaia, Erandio, Bilbao, Spain
| | - Lamia Dahdah
- Translational Research in Pediatric Specialties Area, Division of Allergy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Motohiro Ebisawa
- Clinical Research Center for Allergy and Rheumatology, NHO Sagamihara National Hospital, Kanagawa, Japan
| | - Elena Galli
- Pediatric Allergy Unit, Research Center, San Pietro Hospital - Fatebenefratelli, Rome, Italy
| | - Rose Kamenwa
- Department of Pediatrics and Child Health, Aga Khan University Hospital, Nairobi, Kenya
| | - Gideon Lack
- Department of Women and Children's Health/Peter Gorer Department of Immunobiology, School of Life Course Sciences, Faculty of Life Sciences & Medicine, King's College London, United Kingdom
- Evelina London Children's Hospital, Guy's and St Thomas' Hospital NHS Foundation Trust, United Kingdom
| | - Haiqi Li
- Department of Primary Child Care, Children's Hospital, Chongqing Medical University, China
| | - Ruby Pawankar
- Department of Pediatrics. Nippon Medical School, Tokyo, Japan
| | | | - Gary Wing Kin Wong
- Department of Paediatrics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Martin Bozzola
- Pediatric Allergy and Immunology Section, Dept of Pediatrics, Hospital Británico de Buenos Aires, Buenos Aires, Argentina
| | - Amal Assa'Ad
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | | | - Sami Bahna
- Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, USA
| | - Jonathan Spergel
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, USA
- Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA, USA
| | - Carina Venter
- Section of Allergy and Clinical Immunology, University of Colorado, USA
- Children's Hospital Colorado. Denver, Colorado, USA
| | - Hania Szajewska
- Department of Paediatrics, The Medical University of Warsaw, Warsaw, Poland
| | - Anna H. Nowak-Wegrzyn
- Department of Pediatrics, Hassenfeld Children's Hospital, New York University, Grossman School of Medicine, New York, NY, USA
- Department of Pediatrics, Gastroenterology and Nutrition, Collegium Medicum, University of Warmia and Mazury, Olsztyn, Poland
| | - Yvan Vandenplas
- Vrije Universiteit Brussel (VUB), UZ Brussel, Belgium
- KidZ Health Castle, Brussels, Belgium
| | - Nikolaos G. Papadopoulos
- Allergy Department, 2nd Pediatric Clinic, University of Athens, Athens, Greece
- Lydia Becker Institute, University of Manchester, Manchester, United Kingdom
| | - Susan Waserman
- Department of Medicine, Division of Clinical Immunology and Allergy, McMaster University, Hamilton, Ontario, Canada
| | - Alessandro Fiocchi
- Translational Research in Pediatric Specialties Area, Division of Allergy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Holger J. Schünemann
- Clinical Epidemiology and Research Center (CERC), Humanitas University & Humanitas Research Hospital, Pieve Emanuele, Milano, Italy
| | - Jan L. Brożek
- Department of Health Research Methods, Evidence & Impact, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, Division of Clinical Immunology and Allergy, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
7
|
Azarmi M, Seyed Toutounchi N, Hogenkamp A, Thijssen S, Overbeek SA, Garssen J, Folkerts G, Van't Land B, Braber S. Human Milk Oligosaccharides in Combination with Galacto- and Long-Chain Fructo-Oligosaccharides Enhance Vaccination Efficacy in a Murine Influenza Vaccination Model. Nutrients 2024; 16:2858. [PMID: 39275175 PMCID: PMC11397401 DOI: 10.3390/nu16172858] [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: 08/02/2024] [Revised: 08/23/2024] [Accepted: 08/24/2024] [Indexed: 09/16/2024] Open
Abstract
Early-life nutrition significantly impacts vaccination efficacy in infants, whose immune response to vaccines is weaker compared to adults. This study investigated vaccination efficacy in female C57Bl/6JOlaHsd mice (6 weeks old) fed diets with 0.7% galacto-oligosaccharides (GOS)/long-chain fructo-oligosaccharides (lcFOS) (9:1), 0.3% human milk oligosaccharides (HMOS), or a combination (GFH) for 14 days prior to and during vaccination. Delayed-type hypersensitivity (DTH) was measured by assessing ear swelling following an intradermal challenge. Influvac-specific IgG1 and IgG2a levels were assessed using ELISAs, while splenic T and B lymphocytes were analyzed for frequency and activation via flow cytometry. Additionally, cytokine production was evaluated using murine splenocytes co-cultured with influenza-loaded dendritic cells. Mice on the GFH diet showed a significantly enhanced DTH response (p < 0.05), increased serological IgG1 levels, and a significant rise in memory B lymphocytes (CD27+ B220+ CD19+). GFH-fed mice also exhibited more activated splenic Th1 cells (CD69+ CXCR3+ CD4+) and higher IFN-γ production after ex vivo restimulation (p < 0.05). These findings suggest that GOS/lcFOS and HMOS, particularly in combination, enhance vaccine responses by improving memory B cells, IgG production, and Th1 cell activation, supporting the potential use of these prebiotics in infant formula for better early-life immune development.
Collapse
Affiliation(s)
- Mehrdad Azarmi
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Science (UIPS), Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Negisa Seyed Toutounchi
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Science (UIPS), Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Astrid Hogenkamp
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Science (UIPS), Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Suzan Thijssen
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Science (UIPS), Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Saskia A Overbeek
- Danone Global Research and Innovation Center B.V., 3584 CT Utrecht, The Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Science (UIPS), Utrecht University, 3584 CG Utrecht, The Netherlands
- Danone Global Research and Innovation Center B.V., 3584 CT Utrecht, The Netherlands
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Science (UIPS), Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Belinda Van't Land
- Danone Global Research and Innovation Center B.V., 3584 CT Utrecht, The Netherlands
- Department of Pediatric Immunology, Wilhelmina Children Hospital, University Medical Center Utrecht, 3584 EA Utrecht, The Netherlands
| | - Saskia Braber
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Science (UIPS), Utrecht University, 3584 CG Utrecht, The Netherlands
- Danone Global Research and Innovation Center B.V., 3584 CT Utrecht, The Netherlands
| |
Collapse
|
8
|
Prince N, Peralta Marzal LN, Markidi A, Ahmed S, Adolfs Y, Pasterkamp RJ, Kumar H, Roeselers G, Garssen J, Kraneveld AD, Perez-Pardo P. Prebiotic diet normalizes aberrant immune and behavioral phenotypes in a mouse model of autism spectrum disorder. Acta Pharmacol Sin 2024; 45:1591-1603. [PMID: 38589690 PMCID: PMC11272935 DOI: 10.1038/s41401-024-01268-x] [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: 11/07/2023] [Accepted: 03/19/2024] [Indexed: 04/10/2024] Open
Abstract
Autism spectrum disorder (ASD) is a cluster of neurodevelopmental disorders characterized by deficits in communication and behavior. Increasing evidence suggests that the microbiota-gut-brain axis and the likely related immune imbalance may play a role in the development of this disorder. Gastrointestinal deficits and gut microbiota dysfunction have been linked to the development or severity of autistic behavior. Therefore, treatments that focus on specific diets may improve gastrointestinal function and aberrant behavior in individuals with ASD. In this study, we investigated whether a diet containing specific prebiotic fibers, namely, 3% galacto-oligosaccharide/fructo-oligosaccharide (GOS/FOS; 9:1), can mitigate the adverse effects of in utero exposure to valproic acid (VPA) in mice. Pregnant BALB/cByJ dams were injected with VPA (600 mg/kg, sc.) or phosphate-buffered saline (PBS) on gestational day 11 (G11). Male offspring were divided into four groups: (1) in utero PBS-exposed with a control diet, (2) in utero PBS-exposed with GOS/FOS diet, (3) in utero VPA-exposed with a control diet, and (4) in utero VPA-exposed with GOS/FOS diet. Dietary intervention started from birth and continued throughout the duration of the experiment. We showed that the prebiotic diet normalized VPA-induced alterations in male offspring, including restoration of key microbial taxa, intestinal permeability, peripheral immune homeostasis, reduction of neuroinflammation in the cerebellum, and impairments in social behavior and cognition in mice. Overall, our research provides valuable insights into the gut-brain axis involvement in ASD development. In addition, dietary interventions might correct the disbalance in gut microbiota and immune responses and, ultimately, might improve detrimental behavioral outcomes in ASD.
Collapse
Affiliation(s)
- Naika Prince
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Lucia N Peralta Marzal
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Anastasia Markidi
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
- Division of Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Utrecht University, 3584 CL, Utrecht, The Netherlands
| | - Sabbir Ahmed
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Youri Adolfs
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Himanshu Kumar
- Danone Nutricia Research, 3584 CT, Utrecht, The Netherlands
| | - Guus Roeselers
- Danone Nutricia Research, 3584 CT, Utrecht, The Netherlands
| | - Johan Garssen
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
- Danone Nutricia Research, 3584 CT, Utrecht, The Netherlands
| | - Aletta D Kraneveld
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
- Department of Neuroscience, Faculty of Science, VU university, 1081 HV, Amsterdam, The Netherlands
| | - Paula Perez-Pardo
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands.
| |
Collapse
|
9
|
Klaassens ES, Baak ML, Mekkes NJ, Bongoni R, Schaubeck M. Effect of protein modification in synbiotic infant formula on probiotic metabolic activity and bacterial composition in an infant gut-model. MICROBIOME RESEARCH REPORTS 2024; 3:38. [PMID: 39421252 PMCID: PMC11480727 DOI: 10.20517/mrr.2024.13] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/30/2024] [Accepted: 06/25/2024] [Indexed: 10/19/2024]
Abstract
Aim: Microbial colonization of the neonatal gut is pivotal in priming the infant's immune system. Human milk (HM) is the best nutrition for infants and supports the development of the microbiota due to prebiotic compounds and probiotic microorganisms. When exclusive breastfeeding is not possible, infant formula (IF) with probiotics is a strategy to support the infant's microbiome development. However, knowledge about the effects of the infant gut microbiota and different compounds in IF on individual probiotic strains is limited, as strain-level detection in a complex ecosystem is challenging. The aim of the present study was to show the effects of IF with different protein forms on the metabolic activity of two probiotic strains isolated from HM in a complex ecosystem. Methods: By using an ex-vivo infant gut model containing infant donor-microbiota, the effects of IF with either intact or extensively hydrolyzed protein on the metabolic activity of the donor microbiota, as well as two probiotic strains [Limosilactobacillus fermentum (L. fermentum) CECT 5716 (Lf) and Bifidobacterium breve (B. breve) DSM 32583 (Bb)], were analyzed. A new bioinformatic pipeline combined with a specific infant microbiome database was used to explore shotgun metagenome datasets (1200 Megabases) for taxonomic identification and strain-level tracking. Results: Both protein forms (i.e., intact or extensively hydrolyzed protein) in IF supported infant gut microbial metabolic activity equally, as evidenced by similar levels of short-chain fatty acids (SCFAs). Interestingly, gut microbial metabolic activity was found to be differently activated in a strain-dependent manner. Taxonomic profiling of the microbiome at the strain level enabled monitoring of the prevalence and abundance of both probiotic strains, even in a complex ecosystem. Conclusion: Food matrix and host microbiota interactions should be considered when evaluating strain-specific probiotic effects in the future.
Collapse
Affiliation(s)
| | | | | | | | - Monika Schaubeck
- Research & Development, HiPP GmbH & Co. Vertrieb KG, Pfaffenhofen 85276, Germany
| |
Collapse
|
10
|
Zhang M, Qiao H, Yang S, Kwok LY, Zhang H, Zhang W. Human Breast Milk: The Role of Its Microbiota and Metabolites in Infant Health. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:10665-10678. [PMID: 38691667 DOI: 10.1021/acs.jafc.3c07690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
This review explores the role of microorganisms and metabolites in human breast milk and their impact on neonatal health. Breast milk serves as both a primary source of nutrition for newborns and contributes to the development and maturation of the digestive, immunological, and neurological systems. It has the potential to reduce the risks of infections, allergies, and asthma. As our understanding of the properties of human milk advances, there is growing interest in incorporating its benefits into personalized infant nutrition strategies, particularly in situations in which breastfeeding is not an option. Future infant formula products are expected to emulate the composition and advantages of human milk, aligning with an evolving understanding of infant nutrition. The long-term health implications of human milk are still under investigation.
Collapse
Affiliation(s)
- Meng Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Hui Qiao
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Shuwei Yang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Lai-Yu Kwok
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Heping Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Wenyi Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| |
Collapse
|
11
|
Hu R, Xu J, Hua Y, Li Y, Li J. Could early life DHA supplementation benefit neurodevelopment? A systematic review and meta-analysis. Front Neurol 2024; 15:1295788. [PMID: 38645744 PMCID: PMC11032049 DOI: 10.3389/fneur.2024.1295788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 03/22/2024] [Indexed: 04/23/2024] Open
Abstract
Background Docosahexaenoic acid (DHA) plays a crucial role in the growth and functional development of the infant brain. However, the impact of additional DHA supplementation on neurodevelopment in infants remains controversial in randomized controlled trials. In this systematic review and meta-analysis, we aimed to investigate the effects of prenatal and postnatal DHA supplementation on neurodevelopment. Methods We systematically searched the MEDLINE, EMBASE, and Cochrane Library electronic databases using a predefined strategy until 8 February 2024. We extracted relevant study characteristics and outcomes related to the nervous system. Two independent reviewers critically evaluated the included studies to assess their validity and risk of bias. Results A total of 21 studies met our inclusion criteria, one study was removed after quality assessment, and the meta-analysis included 9 randomized controlled trials. The meta-analysis results indicated that there was no statistically significant difference between the DHA supplementation group and the placebo group, as assessed by the Mental Development Index [MDI; mean difference (MD), 0.41; 95% confidence interval (CI), -0.91 to 1.73; p = 0.55]. However, the DHA group had a significantly higher Psychomotor Development Index (PDI) than the placebo group (MD, 1.47; 95% CI, 0.23 to 2.72; p = 0.02). Subgroup analyses based on populations showed that DHA supplementation was superior to placebo for infants in both MDI (language score conversion; MD, 2.05; 95% CI, -0.16 to 4.26; p = 0.07) and PDI (MD, 1.94; 95% CI, 0.23 to 3.65; p = 0.03). Other subgroup analyses indicated no statistical differences between the two groups. The remaining assessments that could not be summarized quantitatively underwent a narrative evaluation. Conclusion Based on the BSID assessments, DHA supplementation in infants may have potential neurodevelopmental benefits. Because the meta-analysis included few high-quality articles and had some limitations, more relevant articles are needed to address the need for separate DHA supplementation in infants, pregnant women, and lactating mothers. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42022348100, identifier: CRD42022348100.
Collapse
Affiliation(s)
| | | | | | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jinrong Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
12
|
Park J, Wickramasinghe S, Mills DA, Lönnerdal BL, Ji P. Iron Fortification and Inulin Supplementation in Early Infancy: Evaluating the Impact on Iron Metabolism and Trace Mineral Status in a Piglet Model. Curr Dev Nutr 2024; 8:102147. [PMID: 38645881 PMCID: PMC11026733 DOI: 10.1016/j.cdnut.2024.102147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/18/2024] [Accepted: 03/22/2024] [Indexed: 04/23/2024] Open
Abstract
Background Infant formula in the United States contains abundant iron, raising health concerns about excess iron intake in early infancy. Objectives Using a piglet model, we explored the impact of high iron fortification and prebiotic or synbiotic supplementation on iron homeostasis and trace mineral bioavailability. Methods Twenty-four piglets were stratified and randomly assigned to treatments on postnatal day 2. Piglets were individually housed and received an iron-adequate milk diet (AI), a high-iron milk diet (HI), HI supplemented with 5% inulin (HI with a prebiotic [HIP]), or HIP with an oral gavage of Ligilactobacillus agilis YZ050, an inulin-fermenting strain, every third day (HI with synbiotic [HIS]). Milk was provided in 14 meals daily, mimicking formula feeding in infants. Fecal consistency score and body weight were recorded daily or every other day. Blood and feces were sampled weekly, and tissues collected on postnatal day 29. Data were analyzed using mixed model analysis of variance with repeated measures whenever necessary. Results Diet did not affect growth. HI increased hemoglobin, hematocrit, and serum iron compared to AI. Despite marginal adequacy, AI upregulated iron transporter genes and maintained satisfactory iron status in most pigs. HI upregulated hepcidin gene expression in liver, caused pronounced tissue iron deposition, and markedly increased colonic and fecal iron. Inulin supplementation, regardless of L. agilis YZ050, not only attenuated hepatic iron overload but also decreased colonic and fecal iron without altering pH or the expression of iron regulatory genes. HI lowered zinc (Zn) and copper (Cu) in the duodenum and liver compared to AI, whereas HIP and HIS further decreased Zn and Cu in the liver and diminished colonic and fecal trace minerals. Conclusions Early-infancy excessive iron fortification causes iron overload and compromises Zn and Cu absorption. Inulin decreases trace mineral absorption likely by enhancing gut peristalsis and stool frequency.
Collapse
Affiliation(s)
- Jungjae Park
- Department of Nutrition, University of California Davis, CA, United States
| | - Saumya Wickramasinghe
- Department of Food Science and Technology, University of California Davis, CA, United States
| | - David A. Mills
- Department of Food Science and Technology, University of California Davis, CA, United States
| | - Bo L. Lönnerdal
- Department of Nutrition, University of California Davis, CA, United States
| | - Peng Ji
- Department of Nutrition, University of California Davis, CA, United States
| |
Collapse
|
13
|
Al-Nassir NSM, Sakr SS. In Vitro Digestibility Assessment of Whey from Goat and Camel Milk Fermented with Lactobacillus helveticus for Use as a Base in Formulating Follow-On Formula. Foods 2024; 13:570. [PMID: 38397547 PMCID: PMC10887542 DOI: 10.3390/foods13040570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/01/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Follow-on formulas are necessary for newborns that are unable to breastfeed. Thus, the development of formulas more tailored to infants' needs is highly important. Recently, using camel milk, goat milk, and sweet milk whey in the formulation of follow-on formulas has gained researchers' attention. Moreover, developing postbiotic systems to create formulas that mimic human milk, are easy to digest, improve compatibility with an infant's gut, and boost immunity is crucial. Thus, this study aimed to create and assess different formulations using fermented whey from camel and goat milks. The fermentation process involved the use of Lactobacillus helveticus as a probiotic and proteolytic lactic acid bacterium strain. The study monitored the proteolytic activity and antioxidant properties of sweet whey produced from cow, camel, and goat milks during the fermentation process with L. helveticus. Also, three different milk fat blends were recombined using edible vegetable oils (coconut oil, rice bran oil, and canola oil) and then they were used to formulate follow-on formulas with a similar fat composition to human milk. Finally, the prepared formulas were tested for their in vitro digestibility and antioxidant activity before and after digestion. The L. helveticus strain had high proteolytic activity towards whey proteins from all the types of milk used in the study. A fermentation time of 6 h produced a higher proteolytic degree and antioxidant activity than 2 and 4 h of fermentation. No significant differences were observed for proteolytic degree and antioxidant activity between 6 and 12 h of fermentation for the cow, camel, and goat whey samples. Regarding the fat blends, animal milk fat, rice bran oil, and canola oil in a fat combination were essential to provide the required amount of unsaturated fatty acids in the follow-on formulas, especially the linoleic acid-α-linolenic acid (LA:ALA) ratio. Adding coconut oil in small amounts to the follow-on formulas provided the required amounts of saturated fatty acids, especially lauric and meristic acids. The follow-on formula based on cow or goat milk whey fermented with L. helveticus released more free amino acids (mmol tyrosine equivalent mL-1) with high levels of antioxidants compared to unfermented ones. The release of free amino acids in the follow-on formula based on camel milk whey was not affected by fermentation. Our results recommend using L. helveticus in the fermentation of follow-on formulas based on camel and goat whey instead of formulas based on cow milk proteins.
Collapse
Affiliation(s)
- Noura S. M. Al-Nassir
- Department of Food Science and Human Nutrition, College of Agriculture and Veterinary Medicine, Qassim University, Buraydah 51452, Saudi Arabia;
| | - Sally S. Sakr
- Department of Food Science and Human Nutrition, College of Agriculture and Veterinary Medicine, Qassim University, Buraydah 51452, Saudi Arabia;
- Dairy Science Department, Faculty of Agriculture, Cairo University, Giza 12613, Egypt
| |
Collapse
|
14
|
Hörnell A, Lagström H. Infant feeding-a scoping review for Nordic Nutrition Recommendations 2023. Food Nutr Res 2024; 68:10456. [PMID: 38370110 PMCID: PMC10870977 DOI: 10.29219/fnr.v68.10456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 12/11/2022] [Accepted: 12/20/2023] [Indexed: 02/20/2024] Open
Abstract
The 2012 edition of the Nordic Nutrition Recommendations (NNR) included recommendations on breastfeeding, based on the most recent guidelines and recommendations from major national food and health authorities and organizations, systematic reviews, and some original research. For NNR 2023, the scope has been expanded and also includes formula feeding and the introduction of solid food. The main focus in this scoping review is on infants aged 0-12 months but also considers parts both before and beyond the first year, as the concept of 'the first 1000 days' emphasizes the importance of factors during pregnancy and the first 2 years of life for immediate and later health: physical as well as emotional and mental health. Breastmilk is the natural and sustainable way to feed an infant during the first months of life. Numerous studies have indicated immediate as well as long-term beneficial effects of breastfeeding on health for both the infant and the breastfeeding mother, and from a public health perspective, it is therefore important to protect, support, and promote breastfeeding. For full-term, normal weight infants, breastmilk is sufficient as the only form of nutrition for the first 6 months, except for vitamin D that needs to be given as supplement. The World Health Organization (WHO) and several other authoritative bodies therefore recommend exclusive breastfeeding during the first 6 months. Starting solids at about 6 months is necessary for both nutritional and developmental reasons. According to the European Food Safety Authority (EFSA) and the European Society for Paediatric Gastroenterology, Hepatology, and Nutrition (ESPGHAN), solid foods are safe to give from 4 months although exclusive breastfeeding until 6 months is the desirable goal. Breastfeeding can continue together with complementary foods as long as it is mutually desired by the mother and child. If breastfeeding is not enough or for some reason discontinued before the infant is 4 months of age, the infant should be fed infant formula, and, when possible, breastfeeding should be continued alongside the formula feeding. If the infant is 4 months or older, starting with solids together with continued breastfeeding and/or formula feeding is an option. Infant formulas have been developed for infants who are not breastfed or do not get enough breastmilk. Home-made formula should not be given.
Collapse
Affiliation(s)
- Agneta Hörnell
- Department of Food, Nutrition and Culinary Science, Umeå University, Umeå, Sweden
| | - Hanna Lagström
- Department of Public Health, University of Turku and Turku University Hospital, Turku, Finland
| |
Collapse
|
15
|
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
|
16
|
Biagioli V, Volpedo G, Riva A, Mainardi P, Striano P. From Birth to Weaning: A Window of Opportunity for Microbiota. Nutrients 2024; 16:272. [PMID: 38257165 PMCID: PMC10819289 DOI: 10.3390/nu16020272] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/12/2024] [Accepted: 01/14/2024] [Indexed: 01/24/2024] Open
Abstract
(1) Background: The first 1000 days of life constitute a critical window of opportunity for microbiota development. Nutrients play a crucial role in enriching and diversifying the microbiota, derived not only from solid food but also from maternal dietary patterns during gestation. (2) Methods: We conducted a comprehensive literature review using the PubMed database, covering eleven years (2013-2023). We included English-language reviews, original research papers, and meta-analyses, while excluding case reports and letters. (3) Results: Consensus in the literature emphasizes that our interaction with a multitude of microorganisms begins in the intrauterine environment and continues throughout our lives. The existing data suggest that early nutritional education programs, initiated during pregnancy and guiding infant diets during development, may influence the shaping of the gut microbiota, promoting long-term health. (4) Conclusions: Further research is necessary in the coming years to assess potential interventions and early nutritional models aimed at modulating the pediatric microbiota, especially in vulnerable populations such as premature newborns.
Collapse
Affiliation(s)
- Valentina Biagioli
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16126 Genoa, Italy; (G.V.); (A.R.); (P.M.); (P.S.)
| | - Greta Volpedo
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16126 Genoa, Italy; (G.V.); (A.R.); (P.M.); (P.S.)
| | - Antonella Riva
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16126 Genoa, Italy; (G.V.); (A.R.); (P.M.); (P.S.)
| | - Paolo Mainardi
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16126 Genoa, Italy; (G.V.); (A.R.); (P.M.); (P.S.)
| | - Pasquale Striano
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16126 Genoa, Italy; (G.V.); (A.R.); (P.M.); (P.S.)
- IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| |
Collapse
|
17
|
Libertini G. Phenoptosis and the Various Types of Natural Selection. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:2007-2022. [PMID: 38462458 DOI: 10.1134/s0006297923120052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/14/2023] [Accepted: 09/17/2023] [Indexed: 03/12/2024]
Abstract
In the first description of evolution, the fundamental mechanism is the natural selection favoring the individuals best suited for survival and reproduction (selection at the individual level or classical Darwinian selection). However, this is a very reductive description of natural selection that does not consider or explain a long series of known phenomena, including those in which an individual sacrifices or jeopardizes his life on the basis of genetically determined mechanisms (i.e., phenoptosis). In fact, in addition to (i) selection at the individual level, it is essential to consider other types of natural selection such as those concerning: (ii) kin selection and some related forms of group selection; (iii) the interactions between the innumerable species that constitute a holobiont; (iv) the origin of the eukaryotic cell from prokaryotic organisms; (v) the origin of multicellular eukaryotic organisms from unicellular organisms; (vi) eusociality (e.g., in many species of ants, bees, termites); (vii) selection at the level of single genes, or groups of genes; (viii) the interactions between individuals (or more precisely their holobionts) of the innumerable species that make up an ecosystem. These forms of natural selection, which are all effects and not violations of the classical Darwinian selection, also show how concepts as life, species, individual, and phenoptosis are somewhat not entirely defined and somehow arbitrary. Furthermore, the idea of organisms selected on the basis of their survival and reproduction capabilities is intertwined with that of organisms also selected on the basis of their ability to cooperate and interact, even by losing their lives or their distinct identities.
Collapse
Affiliation(s)
- Giacinto Libertini
- Italian Society for Evolutionary Biology (ISEB), Asti, 14100, Italy.
- Department of Translational Medical Sciences, Federico II University of Naples, Naples, 80131, Italy
| |
Collapse
|
18
|
Alves Gragnani Vido M, Dutra Alvim I, Vinderola G, Isabel Berto M, Blumer Zacarchenco Rodrigues de Sá P, Mauricio Barreto Pinilla C, Torres Silva E Alves A. Microencapsulation of Limosilactobacillus reuteri (DSM 23878) for application in infant formula: Heat resistance and bacterial viability during long-time storage. Food Res Int 2023; 173:113378. [PMID: 37803716 DOI: 10.1016/j.foodres.2023.113378] [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: 03/21/2023] [Revised: 08/11/2023] [Accepted: 08/16/2023] [Indexed: 10/08/2023]
Abstract
This study aimed to evaluate the survival capacity of the probiotic culture Limosilactobacillus reuteri (DSM 23878) to microencapsulation by spray drying, and its potential as component of an infant formula. Preliminary tests were performed between skim milk (SM) and infant formula (IF) as wall material and two inlet temperatures, evaluating the encapsulation efficiency, moisture content, water activity and stability, to choose the drying parameters. After drying in optimized conditions, the powder of microencapsulated L. reuteri was characterized and the viability after dilution in an infant formula at 70 °C was determined. In addition, the survival rate throughout 360 days of storage was assessed. As results, encapsulation efficiency was superior to 90 % in both wall materials. However, the use of IF as for microencapsulation produced microparticles with lower water activity (Aw) and moisture, as compared with the SM. Final microparticles produced with IF as wall material presented values of Aw, moisture content, and particle diameter averaged 0.11 ± 0.02, 2.10 ± 0.35 % and 10.30 ± 0.12 μm, respectively. The viability of microencapsulated L.reuteri decreased 1 Log CFU/mL after dilution at 70 °C and the powder maintained a survivor of 73.5 % after 365 days of storage at 4 °C. Thus, the microencapsulation by spray drying under the conditions of this study proved to be an effective technique to protect the probiotic L. reuteri for application in infant formulas, obtaining an adequate number of viable cells after reconstitution at 70 °C and during long time the storage.
Collapse
Affiliation(s)
| | - Izabela Dutra Alvim
- Cereal and Chocolate Technology Center, Institute of Food Technology, Brazil (ITAL), Campinas, São Paulo, Brazil
| | - Gabriel Vinderola
- Instituto de Lactología Industrial (CONICET-UNL), Facultad de Ingeniería Química, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Maria Isabel Berto
- Dairy Technology Center (TECNOLAT) of the Food Technology Institute (ITAL), Campinas, São Paulo, Brazil
| | | | | | | |
Collapse
|
19
|
Soria R, Del Compare M, Sallaberry M, Martín G, Aprigliano G, Hermida V, Carosella M, Gruenberg M, Monsell S, Micone P, Maciero E, Giglio N. Efficacy of an extensively hydrolyzed formula with the addition of synbiotics in infants with cow's milk protein allergy: a real-world evidence study. FRONTIERS IN ALLERGY 2023; 4:1265083. [PMID: 37876766 PMCID: PMC10591099 DOI: 10.3389/falgy.2023.1265083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 09/11/2023] [Indexed: 10/26/2023] Open
Abstract
Introduction Cow's milk protein allergy (CMPA) is the most frequent food allergy in early childhood. For those infants requiring breastmilk substitutes, formulas with extensively hydrolyzed proteins (EHF), should be the treatment of choice. As there are limited data showing the progression of initial symptoms in infants newly diagnosed with CMPA who are treated with EHF with added synbiotics, the main objective of this study was to evaluate the resolution of symptoms in said infants after 4 weeks of treatment. As a secondary objective this study aimed to assess the impact of the treatment on the family's quality of life. Materials and Methods observational, longitudinal, prospective, and multicentric real-world evidence study. The intervention phase (EHF with synbiotics) lasted 28 days and was completed by 65 patients. Treating physicians registered child´s anthropometry, Infant Gastrointestinal Symptoms Questionnaire (IGSQ-13) and CoMiSS (Cow´s Milk Allergy Symptoms Score) both at baseline and after 28 days of treatment. During treatment, caregivers reported child´s regurgitation and stools, PO-SCORAD (Patient Oriented Scoring of Atopic Dermatitis) and FAQL-PB (Family Quality of Life-Parental Burden). Data were collected using Google Forms and analyzed through the STATA program. Results 95.4% of the patients showed an improvement or disappearance of the overall initial symptoms after 4 weeks of treatment. Gastrointestinal symptoms improved or disappeared in 92% of patients (p < 0.05) while dermatological symptoms improved or disappeared in 87.5% of patients (p < 0.05). The median CoMiSS at baseline was 9, with 21 patients exceeding the cut-off point of 12. After 4 weeks of treatment, the median dropped to 3, and no patient exceeded the 12-cut-off point (p = 0.000). At baseline, patients had a PO-SCORAD of 11.5 (interquartile range 1-23) that went to 1.0 (interquartile range 1-6) at day 28 (p = 0.000). The treatment diminished stool frequency (p < 0.05), improved stool consistency (p = 0.004) and decreased the frequency of regurgitation in infants with CMPA (p = 0.01). The percentage of patients who no longer had any episode of regurgitation increased from 11% to 31% on day 28 (p = 0.003). At baseline, 13% of patients cried more than 3 h per day, while at day 28 that percentage dropped to 3% (p = 0.03). An improvement in the infants' sleep pattern was also appreciated with the treatment. At study onset, 56% of the families reported feeling very overwhelmed, a percentage that dropped to 17% after 28 days of treatment (p < 0.05). The small percentage of families who did not feel overwhelmed at study onset (17%), grew to 43% on day 28 (p < 0.05). Conclusions The use of an EHF with synbiotics for the management of infants diagnosed with or suspected to have CMPA suggested a good safety profile, an adequate infant growth, and improvement of overall, gastrointestinal, and dermatological symptoms. It also suggested a lower daily frequency of regurgitations and stools, and an improvement in stool consistency, sleeping pattern, and quality of life of the infant and his family.
Collapse
Affiliation(s)
- Ramiro Soria
- Sanatorio Infantil San Lucas, San Miguel de Tucumán, Tucumán, Argentina
| | - Mónica Del Compare
- Consultorios Externos Sanatorio Mater Dei, Ciudad Autónoma de Buenos Aires, Argentina
| | | | - Graciela Martín
- Clínica del Niño y Consultorios de Especialistas, Corrientes, Argentina
| | - Gustavo Aprigliano
- Consultorio Pediátrico Urquiza, Ciudad Autónoma de Buenos Aires, Argentina
| | - Verónica Hermida
- Consultorios Neuropediatría Barracas, Ciudad Autónoma de Buenos Aires, Argentina
| | - Mabel Carosella
- Grupo Pediátrico BelgranoR, Ciudad Autónoma de Buenos Aires, Argentina
| | | | - Silvana Monsell
- Consultorio Privado, Ciudad Autónoma de Buenos Aires, Argentina
| | - Paula Micone
- Servicio de Tocoginecología, Hospital Durand, Ciudad Autónoma de Buenos Aires, Argentina
| | - Eugenia Maciero
- Grupo Pediátrico BelgranoR, Ciudad Autónoma de Buenos Aires, Argentina
| | - Norberto Giglio
- Grupo Pediátrico BelgranoR, Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
20
|
Bellaiche M, Tounian P, Oozeer R, Rocher E, Vandenplas Y. Digestive Tolerance and Safety of an Anti-Regurgitation Formula Containing Locust Bean Gum, Prebiotics and Postbiotics: A Real-World Study. Pediatr Gastroenterol Hepatol Nutr 2023; 26:249-265. [PMID: 37736220 PMCID: PMC10509020 DOI: 10.5223/pghn.2023.26.5.249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 06/20/2023] [Accepted: 07/20/2023] [Indexed: 09/23/2023] Open
Abstract
Purpose Infant regurgitation is associated with other functional gastrointestinal disorders and signs and symptoms that have a major impact on the quality of life of infants and their families. This study evaluated the safety, tolerance, and real-world effectiveness of an anti-regurgitation formula containing locust bean gum (LBG), prebiotics, and postbiotics to alleviate digestive symptoms beyond regurgitation. Methods This 3-month study involved infants with regurgitation requiring the prescription of an anti-regurgitation formula according to usual clinical practice. Outcomes included evaluation of the evolution of stool consistency and frequency; occurrence of colic, constipation, and diarrhea; and assessment of regurgitation severity. Infant crying, parental assessment of infant well-being, and parental satisfaction with the stool consistency were also evaluated. Results In total, 190 infants (average age: 1.9±1.1 months) were included. After three months, stool frequency and consistency remained within the normal physiological range, with 82.7% of infants passing one or two stools per day and 90.4% passing loose or formed stools. There was no significant increase in the number of infants with diarrhea, whereas a decrease was observed in the number of infants with constipation after 1 month (p=0.001) and with colic after both 1 and 3 months (p<0.001). Regurgitation severity and crying decreased and parental satisfaction with stool consistency, formula acceptability, infant well-being, and sleep quality increased. Monitoring of adverse events did not reveal any safety concerns. Conclusion Formulas containing LBG, prebiotics, and postbiotics were well tolerated and provided an effective strategy for managing infant regurgitation and gastrointestinal discomfort.
Collapse
Affiliation(s)
- Marc Bellaiche
- Department of Gastroenterology and Pediatric Nutrition, Hospital Robert-Debré, AP-HP, Paris, France
| | - Patrick Tounian
- Department of Pediatric Nutrition and Gastroenterology, Trousseau Hospital, AP-HP, Sorbonne Université, Paris, France
| | | | | | - Yvan Vandenplas
- KidZ Health Castle, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
21
|
Schönknecht YB, Moreno Tovar MV, Jensen SR, Parschat K. Clinical Studies on the Supplementation of Manufactured Human Milk Oligosaccharides: A Systematic Review. Nutrients 2023; 15:3622. [PMID: 37630811 PMCID: PMC10458772 DOI: 10.3390/nu15163622] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/12/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Human milk oligosaccharides (HMOs) are a major component of human milk. They are associated with multiple health benefits and are manufactured on a large scale for their addition to different food products. In this systematic review, we evaluate the health outcomes of published clinical trials involving the supplementation of manufactured HMOs. We screened the PubMed database and Cochrane Library, identifying 26 relevant clinical trials and five publications describing follow-up studies. The clinical trials varied in study populations, including healthy term infants, infants with medical indications, children, and adults. They tested eight different HMO structures individually or as blends in varying doses. All trials included safety and tolerance assessments, and some also assessed growth, stool characteristics, infections, gut microbiome composition, microbial metabolites, and biomarkers. The studies consistently found that HMO supplementation was safe and well tolerated. Infant studies reported a shift in outcomes towards those observed in breastfed infants, including stool characteristics, gut microbiome composition, and intestinal immune markers. Beneficial gut health and immune system effects have also been observed in other populations following HMO supplementation. Further clinical trials are needed to substantiate the effects of HMO supplementation on human health and to understand their structure and dose dependency.
Collapse
|
22
|
Sharif S, Oddie SJ, Heath PT, McGuire W. Prebiotics to prevent necrotising enterocolitis in very preterm or very low birth weight infants. Cochrane Database Syst Rev 2023; 6:CD015133. [PMID: 37262358 PMCID: PMC10234253 DOI: 10.1002/14651858.cd015133.pub2] [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] [Indexed: 06/03/2023]
Abstract
BACKGROUND Dietary supplementation with prebiotic oligosaccharides to modulate the intestinal microbiome has been proposed as a strategy to reduce the risk of necrotising enterocolitis (NEC) and associated mortality and morbidity in very preterm or very low birth weight (VLBW) infants. OBJECTIVES To assess the benefits and harms of enteral supplementation with prebiotics (versus placebo or no treatment) for preventing NEC and associated morbidity and mortality in very preterm or VLBW infants. SEARCH METHODS We searched the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, Embase, the Maternity and Infant Care database and the Cumulative Index to Nursing and Allied Health Literature (CINAHL), from the earliest records to July 2022. We searched clinical trials databases and conference proceedings, and examined the reference lists of retrieved articles. SELECTION CRITERIA We included randomised controlled trials (RCTs) and quasi-RCTs comparing prebiotics with placebo or no prebiotics in very preterm (< 32 weeks' gestation) or VLBW (< 1500 g) infants. The primary outcomes were NEC and all-cause mortality, and the secondary outcomes were late-onset invasive infection, duration of hospitalisation since birth, and neurodevelopmental impairment. DATA COLLECTION AND ANALYSIS Two review authors separately evaluated risk of bias of the trials, extracted data, and synthesised effect estimates using risk ratio (RR), risk difference (RD), and mean difference (MD), with associated 95% confidence intervals (CIs). The primary outcomes of interest were NEC and all-cause mortality; our secondary outcome measures were late-onset (> 48 hours after birth) invasive infection, duration of hospitalisation, and neurodevelopmental impairment. We used the GRADE approach to assess the level of certainty of the evidence. MAIN RESULTS We included seven trials in which a total of 705 infants participated. All the trials were small (mean sample size 100). Lack of clarity on methods to conceal allocation and mask caregivers or investigators were potential sources of bias in three of the trials. The studied prebiotics were fructo- and galacto-oligosaccharides, inulin, and lactulose, typically administered daily with enteral feeds during birth hospitalisation. Meta-analyses of data from seven trials (686 infants) suggest that prebiotics may result in little or no difference in NEC (RR 0.97, 95% CI 0.60 to 1.56; RD none fewer per 1000, 95% CI 50 fewer to 40 more; low-certainty evidence), all-cause mortality (RR 0.43, 95% CI 0.20 to 0.92; 40 per 1000 fewer, 95% CI 70 fewer to none fewer; low-certainty evidence), or late-onset invasive infection (RR 0.79, 95% CI 0.60 to 1.06; 50 per 1000 fewer, 95% CI 100 fewer to 10 more; low-certainty evidence) prior to hospital discharge. The certainty of this evidence is low because of concerns about the risk of bias in some trials and the imprecision of the effect size estimates. The data available from one trial provided only very low-certainty evidence about the effect of prebiotics on measures of neurodevelopmental impairment (Bayley Scales of Infant Development (BSID) Mental Development Index score < 85: RR 0.84, 95% CI 0.25 to 2.90; very low-certainty evidence; BSID Psychomotor Development Index score < 85: RR 0.24, 95% 0.03 to 2.00; very low-certainty evidence; cerebral palsy: RR 0.35, 95% CI 0.01 to 8.35; very low-certainty evidence). AUTHORS' CONCLUSIONS The available trial data provide low-certainty evidence about the effects of prebiotics on the risk of NEC, all-cause mortality before discharge, and invasive infection, and very low-certainty evidence about the effect on neurodevelopmental impairment for very preterm or VLBW infants. Our confidence in the effect estimates is limited; the true effects may be substantially different. Large, high-quality trials are needed to provide evidence of sufficient validity to inform policy and practice decisions.
Collapse
Key Words
- humans
- infant, newborn
- enterocolitis, necrotizing
- enterocolitis, necrotizing/etiology
- enterocolitis, necrotizing/prevention & control
- infant, extremely premature
- infant, premature, diseases
- infant, premature, diseases/etiology
- infant, premature, diseases/prevention & control
- infant, very low birth weight
- infections
Collapse
Affiliation(s)
- Sahar Sharif
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Sam J Oddie
- Bradford Neonatology, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Paul T Heath
- Division of Child Health and Vaccine Institute, St. George's, University of London, London, UK
| | - William McGuire
- Centre for Reviews and Dissemination, University of York, York, UK
| |
Collapse
|
23
|
Njayou MM, Ngounouno Ayiwouo M, Ngounouno I. Trace metal contamination status in soils of the abandoned gold mining district of Bindiba (East Cameroon): Pollution indices assessment, multivariate analysis and; geostatistical approach. JOURNAL OF ENVIRONMENTAL HEALTH SCIENCE & ENGINEERING 2023; 21:143-155. [PMID: 37159739 PMCID: PMC10163204 DOI: 10.1007/s40201-023-00849-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 12/18/2022] [Indexed: 05/11/2023]
Abstract
In this study, contamination by trace metals (TMs) such as Cr, Ni, Cu, As, Pb and Sb in the soils of the Bindiba mining district was assessed. This study aims to reveal the current status of the soil quality of the abandoned gold mining district of Bindiba and provide a scientific basis for its future remediation and overall management. 89 soil samples were systematically collected and characterized in order to determine the concentration of TMs (Cr, Ni, Cu, As, Pb and Sb). To assess the degree of metallic contamination, pollution indices were employed. Both multivariate statistical analysis (MSA) and geostatistical modelling (GM) were used to identify the potential sources of TMs elements and to determine the values of the modified contamination degree (mCd), the Nemerow Pollution Index (NPI) and the potential ecological risk index (RI) at un-sampled points. The results of trace metals (TMEs) characterization showed that the concentration of Cr, Ni, Cu, As, Pb and Sb ranged from 22.15-442.44 mg/kg, 9.25-360.37 mg/kg, 1.28-320.86 mg/kg, 0-46.58 mg/kg, 0-53.27 mg/kg and 0-6.33 mg/kg, respectively. The mean concentration of Cr, Cu and Ni exceeds the continental geochemical background values. The Enrichment Factor (EF) assessment indicates two categories of enrichment: moderately to extremely enrichment for Cr, Ni, and Cu and deficiency to minimal enrichment of Pb, As and Sb. Multivariate statistical analysis shows weak linear correlations between the studied heavy metals and suggests that these metals could not come from the same origins. The geostatistical modelling based on the values of mCd, NI and RI suggests a potential high pollution risk existed in the study area. The mCd, NPI and RI interpolation maps showed that the Northern part of the gold mining district was characterized by a high degree of contamination, heavy pollution, and considerable ecological risk. The dispersion of TMs in soils could mainly be attributed to anthropogenic activities and natural phenomena (chemical weathering or erosion). Appropriate measures should be taken to manage and remediate the TMs pollution in this abandoned gold mining district in order to reduce its negative effects on the environment and health of the local population. Supplementary Information The online version contains supplementary material available at 10.1007/s40201-023-00849-y.
Collapse
Affiliation(s)
- Martin Mozer Njayou
- Department of Mining and Geology, School of Geology and Mining Engineering, University of Ngaoundere, P.O. BOX 115, Meiganga, Cameroon
| | - Mouhamed Ngounouno Ayiwouo
- Department of Mining Engineering, School of Geology and Mining Engineering, University of Ngaoundere, P.O. BOX 115, Meiganga, Cameroon
| | - Ismaila Ngounouno
- Department of Earth Sciences, Faculty of Sciences, University of Ngaoundere, P.O. BOX 454, Ngaoundere, Cameroon
| |
Collapse
|
24
|
Cool R, Vandenplas Y. The Link between Different Types of Prebiotics in Infant Formula and Infection Rates: A Review. Nutrients 2023; 15:1942. [PMID: 37111161 PMCID: PMC10140947 DOI: 10.3390/nu15081942] [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: 03/12/2023] [Revised: 04/08/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Breastfeeding plays a protective role against infections, partially through the prebiotic effect of human milk oligosaccharides (HMOs). Aiming to mimic these beneficial capacities, there is an ongoing search to make infant formula closer to human milk, including by adding oligosaccharides. Over the past two decades, multiple studies have been published on different types of prebiotics and their role in reducing infection rates in infants. This review aims to answer the question of whether there is evidence that the addition of oligosaccharides to infant formula decreases the prevalence of infection, and whether the effect is influenced by the kind of oligosaccharide added. The review of the literature reveals an important heterogeneity, including different types and dosages of prebiotics, different intervention periods and inclusion criteria, etc., making it impossible to formulate a consensus about the efficacy of adding prebiotics to infant formula. We would cautiously suggest that supplementation with galactooligosaccharides (GOSs)/fructooligosaccharides (FOSs) seems to have a beneficial effect on infection rates. For HMOs, more studies about the different types of HMOs are necessary to make any deductions. GOSs alone, inulin, and MOSs (bovine-milk-derived oligosaccharides) do not reduce the incidence of infections. The combination of GOSs and PDX (polydextrose) was found to play a protective role in one study. The evidence of the effect of prebiotics in reducing the use of antibiotics is low. The many lacunas in the direction of study uniformity offer many opportunities for further research.
Collapse
Affiliation(s)
| | - Yvan Vandenplas
- Vrije Universiteit Brussel (VUB), UZ Brussel, KidZ Health Castle, 1090 Brussels, Belgium
| |
Collapse
|
25
|
Belyaeva IA, Bombardirova EP, Turti TV. New Strategies for Enhancement of Infant Milk Formulas Composition. CURRENT PEDIATRICS 2022. [DOI: 10.15690/vsp.v21i6.2468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
This article covers the issues of remodeling breast milk’s protective properties during creating infant milk formulas. First of all, this concerns the conditions for normal intestinal microbiota development in growing organism. Its quantitative and qualitative features are the trigger of either sanogenetic, or pathological immune and metabolic reactions, and also determine gut-brain axis functioning. The protective significance of prebiotic composition diversity of mammalian milk and the inductive role of breast milk oligosaccharides are shown. The modern concept of synbiotics role in gastrointestinal tract and other systems functioning, as well as the use of modern synbiotics in the creation of infant formulas (available Russian formula included) are presented.
Collapse
Affiliation(s)
- I. A. Belyaeva
- Research Institute of Pediatrics and Children’s Health in Petrovsky National Research Centre of Surgery; Pirogov Russian National Research Medical University; Morozovskaya Children’s City Hospital
| | - E. P. Bombardirova
- Research Institute of Pediatrics and Children’s Health in Petrovsky National Research Centre of Surgery
| | - T. V. Turti
- Research Institute of Pediatrics and Children’s Health in Petrovsky National Research Centre of Surgery; Pirogov Russian National Research Medical University; Research Institute for Healthcare Organization and Medical Management
| |
Collapse
|
26
|
Belyaeva IA, Namazova-Baranova LS, Bombardirova EP, Turti TV. World Trends in Infant Formulas Composition Enhancement. CURRENT PEDIATRICS 2022. [DOI: 10.15690/vsp.v21i6.2479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
This article provides the overview of the major strategies for infant formulas composition enhancement via modern technologies and trends in minimizing technology-related loads on the environment. Potential modifications of quantitative and qualitative characteristics of milk formulas nutrients have been determined. We also covered product contents changing over age, as well as the perspectives of using animal milk in formulas. The relevance of adding biologically active substances and living microorganisms (probiotics), their safety, and efficacy are discussed.
Collapse
Affiliation(s)
- Irina A. Belyaeva
- Research Institute of Pediatrics and Children’s Health in Petrovsky National Research Centre of Surgery; Pirogov Russian National Research Medical University; Morozovskaya Children’s City Hospital
| | - Leyla S. Namazova-Baranova
- Research Institute of Pediatrics and Children’s Health in Petrovsky National Research Centre of Surgery; Pirogov Russian National Research Medical University
| | - Elena P. Bombardirova
- Research Institute of Pediatrics and Children’s Health in Petrovsky National Research Centre of Surgery
| | - Tatiana V. Turti
- Research Institute of Pediatrics and Children’s Health in Petrovsky National Research Centre of Surgery; Pirogov Russian National Research Medical University; Research Institute for Healthcare Organization and Medical Management
| |
Collapse
|
27
|
Ayechu-Muruzabal V, de Boer M, Blokhuis B, Berends AJ, Garssen J, Kraneveld AD, van’t Land B, Willemsen LEM. Epithelial-derived galectin-9 containing exosomes contribute to the immunomodulatory effects promoted by 2'-fucosyllactose and short-chain galacto- and long-chain fructo-oligosaccharides. Front Immunol 2022; 13:1026031. [PMID: 36685520 PMCID: PMC9846635 DOI: 10.3389/fimmu.2022.1026031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Introduction Early life exposure to non-digestible oligosaccharides (NDO) or microbial components is known to affect immune development. NDO in combination with a TLR9 agonist mimicking bacterial triggers (CpG) promoted the secretion of galectins through unknown pathways. We aimed to study the contribution of exosomes in epithelial galectin secretion and subsequent immunoregulation upon exposure to a mixture of NDO by inhibiting exosome biogenesis. Methods Human intestinal epithelial cells (IEC) (FHs 74 Int or HT-29) were apically exposed to 2'-fucosyllactose (2'FL) and short-chain galacto- and long-chain fructo-oligosaccharides (GF), alone or with CpG. Basolaterally, non-activated or αCD3/CD28-activated peripheral blood mononuclear cells (PBMC) were added. After 24 h incubation, IEC were washed and incubated in fresh medium to analyze epithelial-derived galectin secretion. Additionally, before exposure to NDO and CpG, IEC were exposed to GW4869 to inhibit exosome biogenesis. After 24 h of incubation, IEC were washed and incubated for additional 24 h in the presence of GW4869, after which epithelial-derived galectin secretion was studied. Also, epithelial-derived exosomes were isolated to study the presence of galectins within the exosomes. Results Compared to CpG alone, exposure to 2'FL/GF mixture and CpG, significantly enhanced Th1-type IFNγ, and regulatory IEC-derived galectin-9 secretion in the HT-29/PBMC model. Similarly, in the FHs 74 Int/PBMC co-culture, 2'FL/GF induced immunomodulatory effects in the absence of CpG. Interestingly, galectin-9 and -4 were present in CD63-expressing exosomes isolated from HT-29 supernatants after IEC/PBMC co-culture. Exposure to GW4869 suppressed 2'FL/GF and CpG induced epithelial-derived galectin-9 secretion, which subsequently prevented the rise in IL-10 and reduction in IL-13 secretion observed in the HT-29/PBMC co-culture model upon exposure to 2'FL/GF and CpG. Discussion Exposure to 2'FL/GF and CpG or 2'FL/GF promoted Th1-type regulatory effects in HT-29/PBMC or FHs 74 Int/PBMC co-culture respectively, while Th2-type IL-13 was reduced in association with increased galectin-9 release. Galectin-9 and -4 were present in exosomes from HT-29 and the inhibition of exosome biogenesis inhibited epithelial-derived galectin secretion. This, also affected immunomodulatory effects in IEC/PBMC co-culture suggesting a key role of galectin expressing IEC-derived exosomes in the mucosal immune regulation induced by NDO.
Collapse
Affiliation(s)
- Veronica Ayechu-Muruzabal
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, Netherlands
| | - Merel de Boer
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, Netherlands
| | - Bart Blokhuis
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, Netherlands
| | - Alinda J. Berends
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, Netherlands
- Danone Nutricia Research, Utrecht, Netherlands
| | - Aletta D. Kraneveld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, Netherlands
| | - Belinda van’t Land
- Danone Nutricia Research, Utrecht, Netherlands
- Center for Translational Immunology, The Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, Netherlands
| | - Linette E. M. Willemsen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, Netherlands
| |
Collapse
|
28
|
Youseflu S, Soltani F, Maleki A. The Association of Assisted Reproductive Technology and Breastfeeding Rates: A Systematic Review and Meta-Analysis. Breastfeed Med 2022; 17:841-852. [PMID: 36037058 DOI: 10.1089/bfm.2022.0127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Background: There is an inconsistent result regarding the mode of fertilization and breastfeeding. Objective: This systematic review and meta-analysis aimed to determine the association between the mode of conception and breastfeeding continuity. Materials and Methods: The English electronic databases were searched for up to May 20, 2022. The Newcastle-Ottawa Quality Assessment Scale was used to evaluate the quality of the included studies. Publication bias was assessed using a funnel plot. The heterogeneity of studies was evaluated using I2 statistics. Results of the random-effects meta-analysis were presented using odds ratios (ORs)/relative ratio (RR) and 95% confidence intervals (CIs). Results: A total of 12 articles with 4,929 subjects were included in this systematic review and meta-analysis. The pool estimates of the meta-analysis show that spontaneous conception is associated with breastfeeding continuity up to 6 months (OR/RR = 0.66, 95% CI: 0.46-0.87) and more than 6 months after childbirth (OR/RR = 0.65, 95% CI: 0.32-0.98) compared to the Assisted reproductive technology (ART) conception. The heterogeneity among the studies was high. The level of breastfeeding, plurality, gestational age, and country were confounding factors that affect the heterogeneity of studies. Conclusion: Our findings confirm an inverse association between ART conception and breastfeeding continuity. Due to the importance of breast milk in improving the health of infants who are born in this way, it is recommended to take appropriate interventions in this field. Possible interaction between ART and breastfeeding continuity in different regions is important to point and future studies on this topic were recommended.
Collapse
Affiliation(s)
- Samaneh Youseflu
- Department of Midwifery and Reproductive Health, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Farzaneh Soltani
- Mother and Child Care Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Azam Maleki
- Social Determinants of Health Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
29
|
Urine Metabolomic Profile of Breast- versus Formula-Fed Neonates Using a Synbiotic-Enriched Formula. Int J Mol Sci 2022; 23:ijms231810476. [PMID: 36142388 PMCID: PMC9499619 DOI: 10.3390/ijms231810476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/04/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
The aim of this study was to compare the urine metabolic fingerprint of healthy neonates exclusively breastfed with that of neonates fed with a synbiotic-enriched formula (Rontamil® Complete 1) at four time points (the 3rd and 15th days of life and the 2nd and 3rd months). The determination of urine metabolic fingerprint was performed using NMR metabolomics. Multivariate data analyses were performed with SIMCA-P 15.0 software and R language. Non-distinct profiles for both groups (breastfeeding and synbiotic formula) for the two first time points (3rd and 15th days of life) were detected, whereas after the 2nd month of life, a discrimination trend was observed between the two groups, which was further confirmed at the 3rd month of life. A clear discrimination of the synbiotic formula samples was evident when comparing the metabolites taken in the first days of life (3rd day) with those taken in the 2nd and 3rd months of life. In both cases, OPLS-DA models explained more than 75% of the metabolic variance. Non-distinct metabolomic profiles were obtained between breastfed and synbiotic-formula-fed neonates up to the 15th day of life. Discrimination trends were observed only after the 2nd month of the study, which could be attributed to breastfeeding variations and the consequent dynamic profile of urine metabolites compared to the stable ingredients of the synbiotic formula.
Collapse
|
30
|
Exploring the Potential of Human Milk and Formula Milk on Infants’ Gut and Health. Nutrients 2022; 14:nu14173554. [PMID: 36079814 PMCID: PMC9460722 DOI: 10.3390/nu14173554] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 11/21/2022] Open
Abstract
Early-life gut microbiota plays a role in determining the health and risk of developing diseases in later life. Various perinatal factors have been shown to contribute to the development and establishment of infant gut microbiota. One of the important factors influencing the infant gut microbial colonization and composition is the mode of infant feeding. While infant formula milk has been designed to resemble human milk as much as possible, the gut microbiome of infants who receive formula milk differs from that of infants who are fed human milk. A diverse microbial population in human milk and the microbes seed the infant gut microbiome. Human milk contains nutritional components that promote infant growth and bioactive components, such as human milk oligosaccharides, lactoferrin, and immunoglobulins, which contribute to immunological development. In an attempt to encourage the formation of a healthy gut microbiome comparable to that of a breastfed infant, manufacturers often supplement infant formula with prebiotics or probiotics, which are known to have a bifidogenic effect and can modulate the immune system. This review aims to elucidate the roles of human milk and formula milk on infants’ gut and health.
Collapse
|
31
|
Di Profio E, Magenes VC, Fiore G, Agostinelli M, La Mendola A, Acunzo M, Francavilla R, Indrio F, Bosetti A, D’Auria E, Borghi E, Zuccotti G, Verduci E. Special Diets in Infants and Children and Impact on Gut Microbioma. Nutrients 2022; 14:nu14153198. [PMID: 35956374 PMCID: PMC9370825 DOI: 10.3390/nu14153198] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 07/29/2022] [Accepted: 08/01/2022] [Indexed: 02/07/2023] Open
Abstract
Gut microbiota is a complex system that starts to take shape early in life. Several factors influence the rise of microbial gut colonization, such as term and mode of delivery, exposure to antibiotics, maternal diet, presence of siblings and family members, pets, genetics, local environment, and geographical location. Breastfeeding, complementary feeding, and later dietary patterns during infancy and toddlerhood are major players in the proper development of microbial communities. Nonetheless, if dysbiosis occurs, gut microbiota may remain impaired throughout life, leading to deleterious consequences, such as greater predisposition to non-communicable diseases, more susceptible immune system and altered gut–brain axis. Children with specific diseases (i.e., food allergies, inborn errors of metabolism, celiac disease) need a special formula and later a special diet, excluding certain foods or nutrients. We searched on PubMed/Medline, Scopus and Embase for relevant pediatric studies published over the last twenty years on gut microbiota dietary patterns and excluded case reports or series and letters. The aim of this review is to highlight the changes in the gut microbiota in infants and children fed with special formula or diets for therapeutic requirements and, its potential health implications, with respect to gut microbiota under standard diets.
Collapse
Affiliation(s)
- Elisabetta Di Profio
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, Università di Milano, 20154 Milan, Italy
| | - Vittoria Carlotta Magenes
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, Università di Milano, 20154 Milan, Italy
| | - Giulia Fiore
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, Università di Milano, 20154 Milan, Italy
| | - Marta Agostinelli
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, Università di Milano, 20154 Milan, Italy
| | - Alice La Mendola
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, Università di Milano, 20154 Milan, Italy
| | - Miriam Acunzo
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, Università di Milano, 20154 Milan, Italy
| | - Ruggiero Francavilla
- Pediatric Section, Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70121 Bari, Italy
| | - Flavia Indrio
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Alessandra Bosetti
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, Università di Milano, 20154 Milan, Italy
| | - Enza D’Auria
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, Università di Milano, 20154 Milan, Italy
- Correspondence:
| | - Elisa Borghi
- Department of Health Sciences, University of Milan, 20142 Milan, Italy
| | - Gianvincenzo Zuccotti
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, Università di Milano, 20154 Milan, Italy
- Department of Biomedical and Clinical Sciences L. Sacco, University of Milan, 20144 Milan, Italy
- Pediatric Clinical Research Center, Fondazione Romeo ed Enrica Invernizzi, University of Milan, 20122 Milan, Italy
| | - Elvira Verduci
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, Università di Milano, 20154 Milan, Italy
- Department of Health Sciences, University of Milan, 20142 Milan, Italy
| |
Collapse
|
32
|
Yang X, Jiang S, Deng X, Luo Z, Chen A, Yu R. Effects of Antioxidants in Human Milk on Bronchopulmonary Dysplasia Prevention and Treatment: A Review. Front Nutr 2022; 9:924036. [PMID: 35923207 PMCID: PMC9340220 DOI: 10.3389/fnut.2022.924036] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/21/2022] [Indexed: 12/20/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a severe chronic lung illness that affects neonates, particularly premature infants. It has far-reaching consequences for infant health and their families due to intractable short- and long-term repercussions. Premature infant survival and long-term quality of life are severely harmed by BPD, which is characterized by alveolarization arrest and hypoplasia of pulmonary microvascular cells. BPD can be caused by various factors, with oxidative stress (OS) being the most common. Premature infants frequently require breathing support, which results in a hyperoxic environment in the developing lung and obstructs lung growth. OS can damage the lungs of infants by inducing cell death, inhibiting alveolarization, inducing inflammation, and impairing pulmonary angiogenesis. Therefore, antioxidant therapy for BPD relieves OS and lung injury in preterm newborns. Many antioxidants have been found in human milk, including superoxide dismutase, glutathione peroxidase, glutathione, vitamins, melatonin, short-chain fatty acids, and phytochemicals. Human milk oligosaccharides, milk fat globule membrane, and lactoferrin, all unique to human milk, also have antioxidant properties. Hence, human milk may help prevent OS injury and improve BPD prognosis in premature infants. In this review, we explored the role of OS in the pathophysiology of BPD and related signaling pathways. Furthermore, we examined antioxidants in human milk and how they could play a role in BPD to understand whether human milk could prevent and treat BPD.
Collapse
Affiliation(s)
- Xianpeng Yang
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Shanyu Jiang
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xianhui Deng
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Zichen Luo
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Ailing Chen
- Translational Medicine Laboratory, Research Institute for Reproductive Health and Genetic Diseases, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, China
- *Correspondence: Ailing Chen
| | - Renqiang Yu
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Renqiang Yu
| |
Collapse
|
33
|
Abstract
OBJECTIVES Infant formulas (IF) with postbiotics, defined as inanimate microorganisms and/or their components that confer a health benefit on the host, are available. We systematically updated evidence on the safety and health effects of administering iF with postbiotics (with or without other modifications) compared with standard IF. METHODS The Cochrane Library, MEDLINE, and EMBASE databases were searched to December 2021. RESULTS Eleven randomized controlled trials were included. Using the Cochrane Risk of Bias Tool 2, for the primary outcomes, 5 trials had an overall high risk of bias, and 6 trials had some concerns of bias. Most data were available on IF fermented with Bifidobacterium breve C50 and Streptococcus thermophilus (BB/ST). These formulas, compared with the standard IF, were safe and well tolerated. Postbiotic formulas with additional modifications (ie, formula fermented with BB/ST & prebiotics, partly fermented formula with BB/ST and prebiotics with or without modified milk fat, partly fermented antiregurgitation formula with BB/ST and prebiotics) were generally safe and well tolerated but did not offer clear benefits replicated in other studies. Only limited data were available on formula fermented with Lactobacillus paracasei CBA L74. CONCLUSIONS IF with postbiotics evaluated so far are safe and well tolerated by infants who cannot be breastfed. No firm conclusion can, however, be reached regarding the clinical effects and benefit of one formula over another. It seems reasonable to discuss with healthcare providers current evidence regarding specific modifications in infant formulas and let them decide whether the expected benefits meet expectations and are worth the cost.
Collapse
|
34
|
Vinderola G, Sanders ME, Salminen S. The Concept of Postbiotics. Foods 2022; 11:foods11081077. [PMID: 35454664 PMCID: PMC9027423 DOI: 10.3390/foods11081077] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 03/31/2022] [Accepted: 04/06/2022] [Indexed: 02/07/2023] Open
Abstract
The scientific community has proposed terms such as non-viable probiotics, paraprobiotics, ghostbiotics, heat-inactivated probiotics or, most commonly, postbiotics, to refer to inanimate microorganisms and/or their components that confer health benefits. This article addresses the various characteristics of different definitions of ‘postbiotics’ that have emerged over past years. In 2021, the International Scientific Association for Probiotics and Prebiotics (ISAPP) defined a postbiotic as “a preparation of inanimate microorganisms and/or their components that confers a health benefit on the host”. This definition of postbiotic requires that the whole or components of inactivated microbes be present, with or without metabolic end products. The definition proposed by ISAPP is comprehensive enough to allow the development of postbiotics from different microorganisms, to be applied in different body sites, encouraging innovation in a promising area for any regulatory category and for companion or production animals, and plant or human health. From a technological perspective, probiotic products may contain inanimate microorganisms, which have the potential to impart a health benefit. However, their contribution to health in most cases has not been established, even if at least one probiotic has been shown to confer the same health benefit by live or inanimate cells.
Collapse
Affiliation(s)
- Gabriel Vinderola
- Instituto de Lactología Industrial (INLAIN, UNL-CONICET), Facultad de Ingeniería Química, Universidad Nacional del Litoral, Santa Fe 3000, Argentina
- Correspondence: ; Tel.: +54-9-3426-31-1943
| | - Mary Ellen Sanders
- International Scientific Association for Probiotics and Prebiotics, Centennial, CO 80122, USA;
| | - Seppo Salminen
- Functional Foods Forum, Faculty of Medicine, University of Turku, 20014 Turku, Finland;
| |
Collapse
|
35
|
Yadav M, Kapoor A, Verma A, Ambatipudi K. Functional Significance of Different Milk Constituents in Modulating the Gut Microbiome and Infant Health. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:3929-3947. [PMID: 35324181 DOI: 10.1021/acs.jafc.2c00335] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Human milk, the gold standard for optimal nourishment, controls the microbial composition of infants by either enhancing or limiting bacterial growth. The milk fat globule membrane has gained interest in gut-related functions and cognitive development. The membrane proteins can directly interact with probiotic bacteria, influencing their survival and adhesion through gastrointestinal transit, whereas membrane phospholipids increase the residence time of probiotic bacteria in the gut. The commensal bacteria in milk act as the initial inoculum in building up the gut colonization of an infant, whereas oligosaccharides promote proliferation of beneficial microorganisms. Interestingly, milk extracellular vesicles are also involved in influencing the microbiota composition but are not well-explored. This review highlights the contribution of different milk components in modulating the infant gut microbiota, particularly the fat globule membrane, and the complex interplay between host- and brain-gut microbiota signaling affecting infant and adult health positively.
Collapse
Affiliation(s)
- Monica Yadav
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India
| | - Ayushi Kapoor
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India
| | - Aparna Verma
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India
| | - Kiran Ambatipudi
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India
| |
Collapse
|
36
|
Puntillo M, Segli F, Champagne CP, Raymond Y, Vinderola G. Functional Microbes and Their Incorporation into Foods and Food Supplements: Probiotics and Postbiotics. Annu Rev Food Sci Technol 2022; 13:385-407. [PMID: 35333590 DOI: 10.1146/annurev-food-052720-011545] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Life expectancy has dramatically increased over the past 200 years, but modern life factors such as environmental exposure, antibiotic overuse, C-section deliveries, limited breast-feeding, and diets poor in fibers and microbes could be associated with the rise of noncommunicable diseases such as overweight, obesity, diabetes, food allergies, and colorectal cancer as well as other conditions such as mental disorders. Microbial interventions that range from transplanting a whole undefined microbial community from a healthy gut to an ill one, e.g., so-called fecal microbiota transplantation or vaginal seeding, to the administration of selected well-characterized microbes, either live (probiotics) or not (postbiotics), with efficacy demonstrated in clinical trials, may be effective tools to treat or prevent acute and chronic diseases that humans still face, enhancing the quality of life.
Collapse
Affiliation(s)
- Melisa Puntillo
- Instituto de Lactología Industrial (CONICET-UNL), Facultad de Ingeniería Química, Universidad Nacional del Litoral, Santa Fe, Argentina;
| | - Franco Segli
- Centro de Referencia para Lactobacilos (CERELA-CONICET), Tucumán, Argentina
| | - Claude P Champagne
- Research and Development Centre of Saint-Hyacinthe, Agriculture and Agri-Food Canada, Saint-Hyacinthe, Québec, Canada.,Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Université Laval, Québec, Canada
| | - Yves Raymond
- Research and Development Centre of Saint-Hyacinthe, Agriculture and Agri-Food Canada, Saint-Hyacinthe, Québec, Canada.,Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Université Laval, Québec, Canada
| | - Gabriel Vinderola
- Instituto de Lactología Industrial (CONICET-UNL), Facultad de Ingeniería Química, Universidad Nacional del Litoral, Santa Fe, Argentina;
| |
Collapse
|
37
|
Weighted analysis of 2'-fucosylactose, 3-fucosyllactose, lacto-N-tetraose, 3'-sialyllactose, and 6'-sialyllactose concentrations in human milk. Food Chem Toxicol 2022; 163:112877. [PMID: 35304182 DOI: 10.1016/j.fct.2022.112877] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 02/11/2022] [Accepted: 02/16/2022] [Indexed: 11/22/2022]
Abstract
Over 150 human milk oligosaccharides (HMOs) have been identified and their concentrations in human milk vary depending on Secretor and Lewis blood group status, environmental and geographical factors, lactation stage, gestational period, and maternal health. Quantitation of HMOs in human milk has been the focus of numerous studies, however, comprehensive and weighted statistical analyses of their levels in human milk are lacking. Therefore, weighted means, standard deviations, medians, interquartile ranges, and 90th percentiles for 2'-fucosyllactose (2'-FL), 3-fucosyllactose (3-FL), lacto-N-tetraose (LNT), 3'-sialyllactose (3'-SL) and 6'-sialyllactose (6'-SL) were calculated using random sampling and the levels of these HMOs in human milk reported in the literature. Probability distributions of the reported levels were also constructed. Although the levels reported in the published studies varied, the weighted means for 2'-FL, 3-FL, LNT, 3'-SL, and 6'-SL were calculated to be 2.58, 0.57, 0.94, 0.28, and 0.39 g/L, respectively, which are consistent with those that have been previously determined in other systematic analyses. Likely due to the use of weighting, the 90th percentiles were greater than the 95% confidence limits that have been previously calculated. Our study therefore provides accurate and important statistical data to help support the level of appropriate HMO supplementation in infant formula.
Collapse
|
38
|
Morales-Ferré C, Azagra-Boronat I, Massot-Cladera M, Tims S, Knipping K, Garssen J, Knol J, Franch À, Castell M, Pérez-Cano FJ, Rodríguez-Lagunas MJ. Preventive Effect of a Postbiotic and Prebiotic Mixture in a Rat Model of Early Life Rotavirus Induced-Diarrhea. Nutrients 2022; 14:nu14061163. [PMID: 35334820 PMCID: PMC8954028 DOI: 10.3390/nu14061163] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 02/06/2023] Open
Abstract
Rotavirus (RV) is the main cause of gastroenteritis in children. Prebiotics and, more recently, postbiotics are used for preventing and treating gastrointestinal infections. The aim of this study was to analyze the effects of a LactofidusTM, short-chain galacto-oligosaccharides (scGOS) and long-chain fructo-oligosaccharides (lcFOS) mixture, and their combination on RV infection, in a rat model, for early life diarrhea. Fifteen litters of suckling rats were intragastrically administered daily with the vehicle, the prebiotic mixture, the postbiotic or the combination. The RV was inoculated on day 5 and then fecal samples were clinically evaluated daily. Viral shedding, intestinal permeability assay, in vitro blocking assay, immunoglobulin profiles, and anti-RV response were assessed at day 8 and 16 of life. Cecal microbiota composition, intestinal gene expression, and short chain fatty acids (SCFAs) were analyzed at day 16. The incidence and severity of diarrhea were significantly reduced by all the supplementations. Moreover, they showed blocking activity, changes in the immunoglobulin profiles, in gut microbiota, and in the intestinal gene expression. The prebiotic mixture reduced gut permeability and changed the SCFA profile, whereas the postbiotic enhanced the expression of Toll-like receptors (TLRs). The combination preserved most of the individual observed effects, and furthermore, complementary effects, such as an increase in white blood cells and lymphocytes recruitment, as well as upregulation of TLR7 and TLR9 gene expression.
Collapse
Affiliation(s)
- Carla Morales-Ferré
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Ignasi Azagra-Boronat
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Malén Massot-Cladera
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Sebastian Tims
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
| | - Karen Knipping
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CA Utrecht, The Netherlands
| | - Johan Garssen
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CA Utrecht, The Netherlands
| | - Jan Knol
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
- Laboratory of Microbiology, Wageningen University, 6708 PB Wageningen, The Netherlands
| | - Àngels Franch
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Margarida Castell
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Francisco J. Pérez-Cano
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
- Correspondence: ; Tel.: +34-934-024-505
| | - María J. Rodríguez-Lagunas
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| |
Collapse
|
39
|
Ayechu-Muruzabal V, van de Kaa M, Mukherjee R, Garssen J, Stahl B, Pieters RJ, van’t Land B, Kraneveld AD, Willemsen LEM. Modulation of the Epithelial-Immune Cell Crosstalk and Related Galectin Secretion by DP3-5 Galacto-Oligosaccharides and β-3′Galactosyllactose. Biomolecules 2022; 12:biom12030384. [PMID: 35327576 PMCID: PMC8945669 DOI: 10.3390/biom12030384] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/17/2022] [Accepted: 02/25/2022] [Indexed: 01/27/2023] Open
Abstract
Prebiotic galacto-oligosaccharides (GOS) were shown to support mucosal immune development by enhancing regulatory-type Th1 immune polarization induced by synthetic CpG oligodeoxynucleotides (TLR9 agonist mimicking a bacterial DNA trigger). Epithelial-derived galectin-9 was associated with these immunomodulatory effects. We aimed to identify the most active fractions within GOS based on the degree of polymerization (DP), and to study the immunomodulatory capacities of DP3-sized β-3′galactosyllactose (β-3′GL) using a transwell co-culture model of human intestinal epithelial cells (IEC) and activated peripheral blood mononuclear cells (PBMC). IEC were apically exposed to different DP fractions of GOS or β-3′GL in the presence of CpG, and basolaterally co-cultured with αCD3/CD28-activated PBMC, washed, and incubated in fresh medium for IEC-derived galectin analysis. Only DP3-5 in the presence of CpG enhanced galectin-9 secretion. DP3-sized β-3′GL promoted a regulatory-type Th1 response by increasing IFNγ and IL-10 or galectin-9 concentrations as compared to CpG alone. In addition, IEC-derived galectin-3, -4, and -9 secretion was increased by β-3′GL when combined with CpG. Therefore, the GOS DP3-5 and most effectively DP3-sized β-3′GL supported the immunomodulatory properties induced by CpG by enhancing epithelial-derived galectin secretion, which, in turn, could support mucosal immunity.
Collapse
Affiliation(s)
- Veronica Ayechu-Muruzabal
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CG Utrecht, The Netherlands; (V.A.-M.); (M.v.d.K.); (J.G.); (A.D.K.)
| | - Melanie van de Kaa
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CG Utrecht, The Netherlands; (V.A.-M.); (M.v.d.K.); (J.G.); (A.D.K.)
| | - Reshmi Mukherjee
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CG Utrecht, The Netherlands; (R.M.); (B.S.); (R.J.P.)
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CG Utrecht, The Netherlands; (V.A.-M.); (M.v.d.K.); (J.G.); (A.D.K.)
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands;
| | - Bernd Stahl
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CG Utrecht, The Netherlands; (R.M.); (B.S.); (R.J.P.)
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands;
| | - Roland J. Pieters
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CG Utrecht, The Netherlands; (R.M.); (B.S.); (R.J.P.)
| | - Belinda van’t Land
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands;
- Center for Translational Immunology, The Wilhelmina Children’s Hospital, University Medical Center Utrecht, 3584 EA Utrecht, The Netherlands
| | - Aletta D. Kraneveld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CG Utrecht, The Netherlands; (V.A.-M.); (M.v.d.K.); (J.G.); (A.D.K.)
| | - Linette E. M. Willemsen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CG Utrecht, The Netherlands; (V.A.-M.); (M.v.d.K.); (J.G.); (A.D.K.)
- Correspondence:
| |
Collapse
|
40
|
De Cosmi V, Mazzocchi A, Agostoni C, Visioli F. Fructooligosaccharides: From Breast Milk Components to Potential Supplements. A Systematic Review. Adv Nutr 2022; 13:318-327. [PMID: 34555852 PMCID: PMC8803487 DOI: 10.1093/advances/nmab102] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/12/2021] [Accepted: 08/25/2021] [Indexed: 12/30/2022] Open
Abstract
Breast milk is the optimal food choice for infant growth and development. Among breast milk components, fructooligosaccharides (FOSs) are being actively studied because of their role in microbiota development. In particular, 2'-fucosyllactose is being proposed as a potential supplement/nutraceutical or component of infant formula. In this systematic review, we critically summarize the available information on FOSs and we discuss their future use in infant nutrition. We searched the main electronic databases (PubMed, Embase, and Scopus), with a final check in May 2021. Search terms were inserted individually and using the Boolean tools AND and OR. Relevant articles were identified using the following words: ("fructooligosaccharides" OR "FOS") AND ("human milk" OR "breast milk" OR "donor milk" OR "bank milk"). The search retrieved 1814 articles. After removal of duplicates, we screened 1591 articles based on title, abstract, and exclusive use of the English language. We included articles describing the concentration of FOSs in human milk and assessed the relevant ones. We excluded reviews, studies on animals, and studies exclusively carried out on adults. Also, we excluded studies that have not reported evidence either on FOSs or on galactooligosaccharides from human milk. The resulting publications were reviewed, and 10 studies were included in the systematic review. We conclude that human milk FOSs are, indeed, crucial to infant gut development and their addition to infant formula is safe, well-tolerated, and might provide immune benefits to newborns. However, we would like to underscore the scantiness of human data and the need to avoid the immediate translation of infant research to the commercialization of supplements marketed to adults.
Collapse
Affiliation(s)
- Valentina De Cosmi
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Alessandra Mazzocchi
- Pediatric Intermediate Care Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Carlo Agostoni
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy; Pediatric Intermediate Care Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Francesco Visioli
- Department of Molecular Medicine, University of Padua, Padua, Italy; Madrid Institute for Advanced Studies (IMDEA)-Food, Campus of International Excellence (CEI) Autonomous University of Madrid + Spanish National Research Council (UAM + CSIC), Madrid, Spain.
| |
Collapse
|
41
|
Nogacka AM, Arboleya S, Nikpoor N, Auger J, Salazar N, Cuesta I, Alvarez-Buylla JR, Mantecón L, Solís G, Gueimonde M, Tompkins TA, de los Reyes-Gavilán CG. In Vitro Probiotic Modulation of the Intestinal Microbiota and 2′Fucosyllactose Consumption in Fecal Cultures from Infants at Two Months of Age. Microorganisms 2022; 10:microorganisms10020318. [PMID: 35208773 PMCID: PMC8876326 DOI: 10.3390/microorganisms10020318] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/26/2022] [Accepted: 01/26/2022] [Indexed: 01/17/2023] Open
Abstract
2′-fucosyllactose (2′FL) is one of the most abundant oligosaccharides in human milk, with benefits on neonatal health. Previous results point to the inability of the fecal microbiota from some infants to ferment 2′FL. We evaluated a probiotic formulation, including the strains Lactobacillus helveticus Rosell®-52 (R0052), Bifidobacterium longum subsp. infantis Rosell®-33 (R0033), and Bifidobacterium bifidum Rosell®-71 (R0071), individually or in an 80:10:10 combination on the microbiota and 2′FL degradation. Independent batch fermentations were performed with feces from six full-term infant donors of two months of age (three breastfed and three formula-fed) with added probiotic formulation or the constituent strains in the presence of 2′FL. Microbiota composition was analyzed by 16S rRNA gene sequencing. Gas accumulation, pH decrease and 2′FL consumption, and levels of different metabolites were determined by chromatography. B. bifidum R0071 was the sole microorganism promoting a partial increase of 2′FL degradation during fermentation in fecal cultures of 2′FL slow-degrading donors. However, major changes in microbiota composition and metabolic activity occurred with L. helveticus R0052 or the probiotic formulation in cultures of slow degraders. Further studies are needed to decipher the role of the host intestinal microbiota in the efficacy of these strains.
Collapse
Affiliation(s)
- Alicja M. Nogacka
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (S.A.); (N.S.); (I.C.); (J.R.A.-B.); (M.G.)
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain; (L.M.); (G.S.)
- Correspondence: (A.M.N.); (C.G.d.l.R.-G.); Tel.: +34-985-89-21-31 (A.M.N.)
| | - Silvia Arboleya
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (S.A.); (N.S.); (I.C.); (J.R.A.-B.); (M.G.)
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain; (L.M.); (G.S.)
| | - Naghmeh Nikpoor
- Rosell Institute for Microbiome and Probiotics, Montreal, QC H4P 2R2, Canada; (N.N.); (J.A.); (T.A.T.)
| | - Jeremie Auger
- Rosell Institute for Microbiome and Probiotics, Montreal, QC H4P 2R2, Canada; (N.N.); (J.A.); (T.A.T.)
| | - Nuria Salazar
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (S.A.); (N.S.); (I.C.); (J.R.A.-B.); (M.G.)
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain; (L.M.); (G.S.)
| | - Isabel Cuesta
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (S.A.); (N.S.); (I.C.); (J.R.A.-B.); (M.G.)
| | - Jorge R. Alvarez-Buylla
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (S.A.); (N.S.); (I.C.); (J.R.A.-B.); (M.G.)
| | - Laura Mantecón
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain; (L.M.); (G.S.)
- Pediatrics Service, Central University Hospital of Asturias (HUCA-SESPA), 33011 Oviedo, Asturias, Spain
| | - Gonzalo Solís
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain; (L.M.); (G.S.)
- Pediatrics Service, Central University Hospital of Asturias (HUCA-SESPA), 33011 Oviedo, Asturias, Spain
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (S.A.); (N.S.); (I.C.); (J.R.A.-B.); (M.G.)
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain; (L.M.); (G.S.)
| | - Thomas A. Tompkins
- Rosell Institute for Microbiome and Probiotics, Montreal, QC H4P 2R2, Canada; (N.N.); (J.A.); (T.A.T.)
| | - Clara G. de los Reyes-Gavilán
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (S.A.); (N.S.); (I.C.); (J.R.A.-B.); (M.G.)
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain; (L.M.); (G.S.)
- Correspondence: (A.M.N.); (C.G.d.l.R.-G.); Tel.: +34-985-89-21-31 (A.M.N.)
| |
Collapse
|
42
|
Ferro LE, Crowley LN, Bittinger K, Friedman ES, Decker JE, Russel K, Katz S, Kim JK, Trabulsi J. Effects of prebiotics, probiotics, and synbiotics on the infant gut microbiota and other health outcomes: A systematic review. Crit Rev Food Sci Nutr 2022; 63:5620-5642. [PMID: 37667870 PMCID: PMC10480560 DOI: 10.1080/10408398.2021.2022595] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The primary aim of this review was to systematically evaluate the literature regarding the effect of pre-, pro-, or synbiotic supplementation in infant formula on the gastrointestinal microbiota. The Cochrane methodology for systematic reviews of randomized controlled trials (RCTs) was employed. Five databases were searched and 32 RCTs (2010-2021) were identified for inclusion: 20 prebiotic, 6 probiotic, and 6 synbiotic. The methods utilized to evaluate gastrointestinal microbiota varied across studies and included colony plating, fluorescence in situ hybridization, quantitative real-time polymerase chain reaction, or tagged sequencing of the 16S rRNA gene. Fecal Bifidobacterium levels increased with supplementation of prebiotics and synbiotics but not with probiotics alone. Probiotic and synbiotic supplementation generally increased fecal levels of the bacterial strain supplemented in the formula. Across all pre-, pro-, and synbiotic-supplemented formulas, results were inconsistent regarding fecal Clostridium levels. Fecal pH was lower with some prebiotic and synbiotic supplementation; however, no difference was seen with probiotics. Softer stools were often reported in infants supplemented with pre- and synbiotics, yet results were inconsistent for probiotic-supplemented formula. Limited evidence demonstrates that pre- and synbiotic supplementation increases fecal Bifidobacterium levels. Future studies utilizing comprehensive methodologies and additional studies in probiotics and synbiotics are warranted.
Collapse
Affiliation(s)
- Lynn E. Ferro
- Department of Behavioral Health and Nutrition, University of Delaware, STAR Tower, 100 Discovery Blvd., Newark, DE 19713, USA
| | - Liana N. Crowley
- Department of Behavioral Health and Nutrition, University of Delaware, STAR Tower, 100 Discovery Blvd., Newark, DE 19713, USA
| | - Kyle Bittinger
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Elliot S. Friedman
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jessica E. Decker
- Department of Behavioral Health and Nutrition, University of Delaware, STAR Tower, 100 Discovery Blvd., Newark, DE 19713, USA
| | - Kathryn Russel
- Metropolitan Area Neighborhood Nutrition Alliance, 420 N 20 Street, Philadelphia, PA 19130, USA
| | - Sarah Katz
- Reference and Instructional Services Department, University of Delaware, 181 S. College Avenue, Newark, DE 19717, USA
| | - Jae Kyeom Kim
- Department of Behavioral Health and Nutrition, University of Delaware, STAR Tower, 100 Discovery Blvd., Newark, DE 19713, USA
| | - Jillian Trabulsi
- Department of Behavioral Health and Nutrition, University of Delaware, STAR Tower, 100 Discovery Blvd., Newark, DE 19713, USA
| |
Collapse
|
43
|
Bellaiche M, Ludwig T, Arciszewska M, Bongers A, Gomes C, Świat A, Dakhlia F, Piollet A, Oozeer R, Vandenplas Y. Safety and Tolerance of a Novel Anti-Regurgitation Formula: A Double-Blind, Randomized, Controlled Trial. J Pediatr Gastroenterol Nutr 2021; 73:579-585. [PMID: 34417399 PMCID: PMC8528134 DOI: 10.1097/mpg.0000000000003289] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 05/29/2021] [Indexed: 01/02/2023]
Abstract
OBJECTIVES A novel anti-regurgitation (AR) formula has been designed to support gut health and improve gastrointestinal (GI) symptoms beyond regurgitation. This study assessed the tolerance and safety of this new AR formula. METHODS This was a 4-week double-blind, randomized, controlled trial with a 4-week extension in formula-fed infants with regurgitation. The new AR (Test) formula contained 0.4 g/100 mL locust bean gum (LBG) as thickener, partly fermented formula with postbiotics, and short-chain galacto-oligosaccharides (scGOS) and long-chain fructo-oligosaccharides (lcFOS) (0.4 g/100 mL, ratio 9:1). The Control AR formula contained LBG (0.4 g/100 mL) with postbiotics and has a history of safe use. The primary outcome was the Infant Gastrointestinal Symptom Questionnaire (IGSQ) sum score including stooling, spitting-up/vomiting, crying, fussiness and flatulence. RESULTS All 182 infants screened were enrolled in the study. The primary analysis showed the equivalence of the IGSQ sum scores at Week 4 between groups. IGSQ sum scores improved significantly within 1 week (Mixed Model Repeated Measurement [MMRM], P < 0.001). Post-hoc analyses showed a bigger improvement of the IGSQ score in the Test (n = 38) versus Control (n = 44) group (MMRM, P = 0.008) in infants with more severe gastrointestinal (GI) symptoms (IGSQ score ≥35). Stool characteristics were comparable between groups. Growth related z scores were in line with the WHO child growth standards and both groups showed improvement of regurgitation. Adverse events did not show any safety concerns. CONCLUSIONS The novel AR formula combining LBG, scGOS/lcFOS and postbiotics is well-tolerated, safe and supports adequate growth during the intervention. Post-hoc analyses suggest that the formula results in more improvement of GI symptom burden in infants with more severe symptoms.
Collapse
Affiliation(s)
| | | | | | - Anke Bongers
- Danone Nutricia Research, Utrecht, The Netherlands
| | | | - Agnieszka Świat
- Centrum Medyczne Promed
- Centrum Medyczne Plejady, Krakow, Poland
| | - Faouzi Dakhlia
- Centre Condorcet, Hôpital Privé d’Antony, Antony, France
| | | | - Raish Oozeer
- Danone Nutricia Research, Utrecht, The Netherlands
| | - Yvan Vandenplas
- KidZ Health Castle, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
44
|
Abstract
Microbes in the 21st century are understood as symbionts ‘completing’ the human ‘superorganism’ (Homo sapiens plus microbial partners-in-health). This paper addresses a significant paradox: despite the vast majority of our genes being microbial, the lack of routine safety testing for the microbiome has led to unintended collateral side effects from pharmaceuticals that can damage the microbiome and inhibit innate ‘colonization resistance’ against pathobionts. Examples are discussed in which a Microbiome First Medicine approach provides opportunities to ‘manage our microbes’ holistically, repair dysbiotic superorganisms, and restore health and resilience in the gut and throughout the body: namely, managing nosocomial infections for Clostridioides difficile and Staphylococcus aureus and managing the gut and neural systems (gut–brain axis) in autism spectrum disorder. We then introduce a risk analysis tool: the evidence map. This ‘mapping’ tool was recently applied by us to evaluate evidence for benefits, risks, and uncertainties pertaining to the breastmilk ecosystem. Here, we discuss the potential role of the evidence map as a risk analysis methodology to guide scientific and societal efforts to: (1) enhance ecosystem resilience, (2) ‘manage our microbes’, and (3) minimize the adverse effects of both acute and chronic diseases.
Collapse
|
45
|
Sitorus NL, Dilantika C, Basrowi RW. Perspective of Indonesian Pediatricians on the Role of PrebioticSupplemented Formula towards Immunity, Growth and Development in Preterm Infants: A Preliminary Data. AMERTA NUTRITION 2021. [DOI: 10.20473/amnt.v5i1sp.2021.34-42] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
ABSTRACT Background: Immature immune system in preterm infants is associated with gut dysbiosis and poses significant health risks to their growth and development. Current guidelines for managing preterm infants focuses solely on macro- and micronutrients, whereas preterm infants’ gastrointestinal system requires optimalization to support nutrient absorption. Studies on the positive impacts of prebiotics as supplements have been conducted, but has not been implemented in Indonesia. Indonesian pediatricians’ perspective on these findings needs to be assessed. Objectives: To describe the perspectives of Indonesian pediatricians on the role of gut microbiota balance in supporting immunity, growth, and development of preterm infants, and the role of breastmilk and prebiotic-supplemented formula in optimizing gut microbiota balance. Methods: A cross-sectional study was conducted on 114 Indonesian pediatricians using a previously-validated and previously-used questionnaire on the role of gut microbiota balance on preterm infants, as well as the role of breastmilk and prebiotic-supplemented formula in optimizing gut microbiota balance. Results: Most respondents agreed that gut microbiota balance supports immunity, growth, and development of preterm infants. Respondents also agreed that breastmilk contains nutrients that support gut microbiota balance and when breastmilk becomes unavailable, prebiotic-supplemented formula can be given as substitute. Conclusions: Indonesian pediatricians considered gut microbiota balance to be important for immunity, growth, and development of preterm infants, and breastmilk to be the most ideal source of nutrition for preterm infants in optimizing gut microbiota balance. When breastmilk is unavailable, prebiotic-supplemented formula can be considered as an alternative.
Collapse
|
46
|
Murphy K, Ross RP, Ryan CA, Dempsey EM, Stanton C. Probiotics, Prebiotics, and Synbiotics for the Prevention of Necrotizing Enterocolitis. Front Nutr 2021; 8:667188. [PMID: 34557508 PMCID: PMC8453148 DOI: 10.3389/fnut.2021.667188] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 08/09/2021] [Indexed: 02/02/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a major cause of morbidity and mortality in preterm infants. The exact mechanism by which NEC develops is poorly understood however there is growing evidence to suggest that perturbations in the early-life gut microbiota composition increase the risk for NEC. Modulation of the gut microbiota with probiotics, prebiotics, or in combination (synbiotics) is an area which has attracted intense interest in recent years. In this narrative review, we present an overview of the role of the gut microbiota in the pathogenesis of NEC. We also examine the evidence currently available from randomized controlled trials, observational studies, systematic reviews, and meta-analysis examining the role of probiotics, prebiotics, and synbiotics in reducing the risk of or preventing NEC. Current clinical practice guidelines with recommendations on the routine administration of probiotics to preterm infants for NEC are also explored.
Collapse
Affiliation(s)
- Kiera Murphy
- Food Biosciences Department, Teagasc Food Research Centre, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - R Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - C Anthony Ryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Neonatal Intensive Care Unit, Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
| | - Eugene M Dempsey
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Neonatal Intensive Care Unit, Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
| | - Catherine Stanton
- Food Biosciences Department, Teagasc Food Research Centre, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
47
|
Toutounchi NS, Braber S, Hogenkamp A, Varasteh S, Cai Y, Wehkamp T, Tims S, Leusink-Muis T, van Ark I, Wiertsema S, Stahl B, Kraneveld AD, Garssen J, Folkerts G, van’t Land B. Human Milk Oligosaccharide 3'-GL Improves Influenza-Specific Vaccination Responsiveness and Immunity after Deoxynivalenol Exposure in Preclinical Models. Nutrients 2021; 13:3190. [PMID: 34579070 PMCID: PMC8466816 DOI: 10.3390/nu13093190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/03/2021] [Accepted: 09/10/2021] [Indexed: 01/15/2023] Open
Abstract
Deoxynivalenol (DON), a highly prevalent mycotoxin food contaminant, is known to have immunotoxic effects. In the current study, the potential of dietary interventions with specific mixtures of trans-galactosyl-oligosaccharides (TOS) to alleviate these effects were assessed in a murine influenza vaccination model. Vaccine-specific immune responses were measured in C57Bl/6JOlaHsd mice fed diets containing DON, TOS or a combination, starting 2 weeks before the first vaccination. The direct effects of TOS and its main oligosaccharide, 3'-galactosyl-lactose (3'-GL), on DON-induced damage were studied in Caco-2 cells, as an in vitro model of the intestinal epithelial barrier. Exposure to DON significantly reduced vaccine-specific immune responses and the percentages of Tbet+ Th1 cells and B cells in the spleen. DON significantly altered epithelial structure and integrity in the ileum and reduced the SCFA levels in the cecum. Adding TOS into DON-containing diets significantly improved vaccine-specific immune responses, restored the immune cell balance in the spleen and increased SCFA concentrations in the cecum. Incubating Caco-2 cells with TOS and 3'-GL in vitro further confirmed their protective effects against DON-induced barrier disruption, supporting immune modulation. Overall, dietary intervention with TOS can attenuate the adverse effects of DON on Th1-mediated immune responses and gut homeostasis. These beneficial properties might be linked to the high levels of 3'-GL in TOS.
Collapse
Affiliation(s)
- Negisa Seyed Toutounchi
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (N.S.T.); (S.B.); (A.H.); (S.V.); (Y.C.); (T.L.-M.); (I.v.A.); (B.S.); (A.D.K.); (J.G.); (G.F.)
| | - Saskia Braber
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (N.S.T.); (S.B.); (A.H.); (S.V.); (Y.C.); (T.L.-M.); (I.v.A.); (B.S.); (A.D.K.); (J.G.); (G.F.)
| | - Astrid Hogenkamp
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (N.S.T.); (S.B.); (A.H.); (S.V.); (Y.C.); (T.L.-M.); (I.v.A.); (B.S.); (A.D.K.); (J.G.); (G.F.)
| | - Soheil Varasteh
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (N.S.T.); (S.B.); (A.H.); (S.V.); (Y.C.); (T.L.-M.); (I.v.A.); (B.S.); (A.D.K.); (J.G.); (G.F.)
| | - Yang Cai
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (N.S.T.); (S.B.); (A.H.); (S.V.); (Y.C.); (T.L.-M.); (I.v.A.); (B.S.); (A.D.K.); (J.G.); (G.F.)
| | - Tjalling Wehkamp
- Danone Nutricia Research, 3584 CG Utrecht, The Netherlands; (T.W.); (S.T.); (S.W.)
| | - Sebastian Tims
- Danone Nutricia Research, 3584 CG Utrecht, The Netherlands; (T.W.); (S.T.); (S.W.)
| | - Thea Leusink-Muis
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (N.S.T.); (S.B.); (A.H.); (S.V.); (Y.C.); (T.L.-M.); (I.v.A.); (B.S.); (A.D.K.); (J.G.); (G.F.)
| | - Ingrid van Ark
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (N.S.T.); (S.B.); (A.H.); (S.V.); (Y.C.); (T.L.-M.); (I.v.A.); (B.S.); (A.D.K.); (J.G.); (G.F.)
| | - Selma Wiertsema
- Danone Nutricia Research, 3584 CG Utrecht, The Netherlands; (T.W.); (S.T.); (S.W.)
| | - Bernd Stahl
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (N.S.T.); (S.B.); (A.H.); (S.V.); (Y.C.); (T.L.-M.); (I.v.A.); (B.S.); (A.D.K.); (J.G.); (G.F.)
- Danone Nutricia Research, 3584 CG Utrecht, The Netherlands; (T.W.); (S.T.); (S.W.)
| | - Aletta D. Kraneveld
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (N.S.T.); (S.B.); (A.H.); (S.V.); (Y.C.); (T.L.-M.); (I.v.A.); (B.S.); (A.D.K.); (J.G.); (G.F.)
| | - Johan Garssen
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (N.S.T.); (S.B.); (A.H.); (S.V.); (Y.C.); (T.L.-M.); (I.v.A.); (B.S.); (A.D.K.); (J.G.); (G.F.)
- Danone Nutricia Research, 3584 CG Utrecht, The Netherlands; (T.W.); (S.T.); (S.W.)
| | - Gert Folkerts
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (N.S.T.); (S.B.); (A.H.); (S.V.); (Y.C.); (T.L.-M.); (I.v.A.); (B.S.); (A.D.K.); (J.G.); (G.F.)
| | - Belinda van’t Land
- Danone Nutricia Research, 3584 CG Utrecht, The Netherlands; (T.W.); (S.T.); (S.W.)
- Center of Translational Immunology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands
| |
Collapse
|
48
|
Morales-Ferré C, Azagra-Boronat I, Massot-Cladera M, Tims S, Knipping K, Garssen J, Knol J, Franch À, Castell M, Rodríguez-Lagunas MJ, Pérez-Cano FJ. Effects of a Postbiotic and Prebiotic Mixture on Suckling Rats' Microbiota and Immunity. Nutrients 2021; 13:2975. [PMID: 34578853 PMCID: PMC8469903 DOI: 10.3390/nu13092975] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 01/04/2023] Open
Abstract
Human milk serves as a model for infant formula providing nutritional solutions for infants not able to receive enough mother's milk. Infant formulas aim to mimic the composition and functionality of human milk by providing ingredients reflecting those of the latest human milk insights, such as prebiotics, probiotics and postbiotics. The aim of this study was to examine the effects of the supplementation with a postbiotic (LactofidusTM) and its combination with the prebiotics short-chain galactooligosaccharides (scGOS) and long-chain fructooligosaccharides (lcFOS) in a preclinical model of healthy suckling rats. Pups were supplemented daily with LactofidusTM (POST group) and/or scGOS/lcFOS (P+P and PRE groups, respectively). Body weight and fecal consistency were analyzed. At the end of the study, immunoglobulin (Ig) profile, intestinal gene expression, microbiota composition and short chain fatty acid (SCFA) proportion were quantified. The supplementation with all nutritional interventions modulated the Ig profile, but the prebiotic mixture and the postbiotic induced differential effects: whereas scGOS/lcFOS induced softer feces and modulated microbiota composition and SCFA profile, Lactofidus™ upregulated Toll-like receptors gene expression. The use of the combination of scGOS/lcFOS and Lactofidus™ showed the effects observed for the oligosaccharides separately, as well as showing a synergistic impact on animal growth. Thus, the combined use of both products seems to be a good strategy to modulate immune and microbial features in early life.
Collapse
Affiliation(s)
- Carla Morales-Ferré
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Ignasi Azagra-Boronat
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Malén Massot-Cladera
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Sebastian Tims
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
| | - Karen Knipping
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CA Utrecht, The Netherlands
| | - Johan Garssen
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CA Utrecht, The Netherlands
| | - Jan Knol
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
- Laboratory of Microbiology, Wageningen University, 6708 PB Wageningen, The Netherlands
| | - Àngels Franch
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Margarida Castell
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - María J. Rodríguez-Lagunas
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Francisco J. Pérez-Cano
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| |
Collapse
|
49
|
Sharif S, Oddie SJ, Heath PT, McGuire W. Prebiotics to prevent necrotising enterocolitis in very preterm or very low birth weight infants. Hippokratia 2021. [DOI: 10.1002/14651858.cd015133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Sahar Sharif
- Centre for Reviews and Dissemination; University of York; York UK
| | - Sam J Oddie
- Bradford Neonatology; Bradford Teaching Hospitals NHS Foundation Trust; Bradford UK
| | - Paul T Heath
- Division of Child Health and Vaccine Institute; St. George's, University of London; London UK
| | - William McGuire
- Centre for Reviews and Dissemination; University of York; York UK
| |
Collapse
|
50
|
Verkhnyatskaya SA, Kong C, Klostermann CE, Schols HA, de Vos P, Walvoort MTC. Digestion, fermentation, and pathogen anti-adhesive properties of the hMO-mimic di-fucosyl-β-cyclodextrin. Food Funct 2021; 12:5018-5026. [PMID: 33954318 PMCID: PMC8185958 DOI: 10.1039/d1fo00830g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 04/23/2021] [Indexed: 12/28/2022]
Abstract
Human milk is widely acknowledged as the best food for infants, and that is not just because of nutritional features. Human milk also contains a plethora of bioactive molecules, including a large set of human milk oligosaccharides (hMOs). Especially fucosylated hMOs have received attention for their anti-adhesive effects on pathogens, preventing attachment to the intestine and infection. Because hMOs are generally challenging to produce in sufficient quantities to study and ultimately apply in (medical) infant formula, novel compounds that are inspired by hMO structures (so-called "mimics") are interesting compounds to produce and evaluate for their biological effects. Here we present our thorough study into the digestion, fermentation and anti-adhesive capacity of the novel compound di-fucosyl-β-cyclodextrin (DFβCD), which was inspired by the molecular structures of hMOs. We establish that DFβCD is not digested by α-amylase and also resistant to fermentation by microbial enzymes from a 9 month-old infant inoculum. In addition, we reveal that DFβCD blocks adhesion of enterotoxigenic E. coli (ETEC) to Caco-2 cells, especially when DFβCD is pre-incubated with ETEC prior to addition to the Caco-2 cells. This suggests that DFβCD functions through a decoy effect. We expect that our results inspire the generation and biological evaluation of other fucosylated hMOs and mimics, to obtain a comprehensive overview of the anti-adhesive power of fucosylated glycans.
Collapse
Affiliation(s)
| | - Chunli Kong
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, the Netherlands
| | - Cynthia E Klostermann
- Biobased Chemistry and Technology, Wageningen University & Research, Wageningen, the Netherlands
| | - Henk A Schols
- Laboratory of Food Chemistry, Wageningen University & Research, Wageningen, the Netherlands
| | - Paul de Vos
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, the Netherlands
| | - Marthe T C Walvoort
- Stratingh Institute for Chemistry, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|