1
|
Shafaeizadeh S, Henry CJ, van Helvoort A, Alles M, Abrahamse-Berkeveld M. Tailored recommendations for infant milk formula intake results in more accurate feeding. Eur J Pediatr 2024; 183:4693-4704. [PMID: 39186085 PMCID: PMC11473556 DOI: 10.1007/s00431-024-05726-w] [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: 02/15/2024] [Revised: 08/04/2024] [Accepted: 08/08/2024] [Indexed: 08/27/2024]
Abstract
Currently available guidelines on the daily formula milk requirements of infants are based on the needs of infants with their growth pattern following the 50th percentile of the weight-for-age growth curve. Hence, current recommendations may not thoroughly detail the needs of infants across the broad spectrum of body weight percentiles. This study aimed to provide stratified recommendations for daily formula milk intake of fully formula-fed infants, across different weight-for-age categories from 0 to 4 months. At first, theoretical age- and gender-specific weight ranges were constructed for infants across five pre-defined weight-for-length percentile categories of the WHO growth standard. Thereafter, total daily energy requirements for each category were calculated and converted to daily formula milk needs. Subsequently, these stratified age- and weight-formula milk recommendations were compared to actual daily and relative formula milk of infants in these categories, retrieved from pooled individual infant formula milk intake data derived from 13 clinical intervention trials. A fitted regression model was used to evaluate differences in volume intakes across body weight categories as well as between theoretically derived and actual intake values. Median daily formula milk volume intake (ml/day) of infants differed significantly across the increasing weight-for-age categories at each time point, with significant differences between small and large infants. Interestingly, the relative daily formula milk volume intake (ml/kg/day) was higher for smaller infants compared to larger infants. The mean daily and relative formula milk intakes demonstrated the same pattern based on theoretical calculations as well as for the actual formula milk intake values retrieved from 13 pooled clinical intervention trials. CONCLUSIONS Based on theoretical calculations and actual formula intake data, we conclude that larger infants require a significantly higher daily formula milk intake than smaller infants, and we postulate that infants could benefit from more tailored formula milk intake recommendations. WHAT IS KNOWN • Adequate energy intake during the infancy period is crucial to support optimal growth and organ development, with the potential for long-lasting health effects. • Current available guidelines on the daily formula milk requirements of infants are based on the needs of infants with their growth pattern following the 50th percentile of the weight-for-age growth curve. WHAT IS NEW • Based on using both theoretical calculations and actual formula intake data, larger infants require a significantly higher daily formula milk intake than smaller infants. • Exclusive formula-fed infants could benefit from more tailored formula milk intake recommendations, in early infancy.
Collapse
Affiliation(s)
- Shila Shafaeizadeh
- Danone Nutricia Research, Uppsalalaan 12, 3584 CT, Utrecht, The Netherlands.
| | - Christiani Jeyakumar Henry
- Clinical Nutrition Research Centre (CNRC), Singapore Institute of Food and Biotechnology Innovation (SIFBI), Singapore, Singapore
| | - Ardy van Helvoort
- Danone Nutricia Research, Uppsalalaan 12, 3584 CT, Utrecht, The Netherlands
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Martine Alles
- Danone Nutricia Research, Uppsalalaan 12, 3584 CT, Utrecht, The Netherlands
| | | |
Collapse
|
2
|
Qian J, Fang Z, Chang S, Zeng Z, Zhang J. Effectiveness and safety study of formula containing probiotics, prebiotics, synbiotics on fullterm infants' growth - a systematic review and meta-analysis of randomized controlled study. Eur J Clin Nutr 2024:10.1038/s41430-024-01506-9. [PMID: 39448812 DOI: 10.1038/s41430-024-01506-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND AND OBJECTIVE Probiotics, prebiotics, and synbiotics, are hot topics of research and have been shown to improve the body's disease state and promote health. Analysis of whether infant formula containing probiotcs, prebiotics, synbiotics is beneficial to infant and child growth. METHODS We systematically searched multiple electronic databases (PubMed, Web of Science, The Cochrane Library, Embase) to identify eligible studies published from 1966 to December 25, 2022. Included studies were randomized controlled trials (RCTs) studying the influence of milk powder containing probiotcs, prebiotics, synbiotics on infants and children's growth. RevMan 5.4 was used to analyze the data. RESULTS A total of 55 RCTs with a total sample size of 8868 participants met the inclusion criteria. Milk powder with probiotics, prebiotics, synbiotics does not significantly improve the growth of infants and children (Weight, height, BMI, and Head Circumference); The incidence of minor adverse events (OR 0.88, 95% CI 0.70-1.11 P = 0.28) and serious adverse events (OR 0.92, 95% CI 0.62-1.36 P = 0.67) was also comparable to the control group; The intestinal microbial diversity of infants consuming probiotcs, prebiotics, synbiotics supplemented formula was lower than that of infants consuming formula without probiotcs, prebiotics, synbiotics (SMD -0.88, 95% CI -1.66- -0.1 P = 0.03), but the abundance of individual beneficial flora was increased. (SMD 1.62, 95%CI 0.61-2.62 P = 0.002). In particular, the abundance of Lactobacillus (SMD 1.62, 95% CI 0.61-2.62 P = 0.002). For metabolites, synbiotics increased fecal antibody concentrations (SMD 0.47, 95% CI 0.08-0.86 P = 0.02), but fecal short-chain fatty acid concentrations remained balanced in both groups (SMD 0.05 95% CI -0.17-0.28 P = 0.64). Compared to the control group, infants who consumed formula with prebiotics had softer stools (SMD -1.47, 95% CI -2.23 to -0.7 P = 0.002) and lower stool pH (SMD -0.82, 95% CI -1.15- -0.5 P < 0.00001), there is also more frequency of bowel movements (SMD 0.27, 95% CI 0.09-0.44 P = 0.002). CONCLUSIONS Probiotcs, prebiotics, synbiotics supplemented formulas significantly increased abundance of individual probiotics, alter intestinal antibody secretion, and improve bowel movements. Incidence of adverse reactions did not differ between the two groups. So we can choose formula-supplemented probiotcs, prebiotics, synbiotics to maintain the intestinal health of infants.
Collapse
Affiliation(s)
- Jiafen Qian
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Zongwei Fang
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Sijie Chang
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Zhiwei Zeng
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Jinhua Zhang
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
3
|
Vray M, Tondeur L, Hedible BG, Randremanana RV, Manirakiza A, Lazoumar RH, Platen CV, Vargas A, Briend A, Jambou R. Three-arm clinical trial of improved flour targeting intestinal microbiota (MALINEA). MATERNAL & CHILD NUTRITION 2024; 20:e13649. [PMID: 38599819 PMCID: PMC11168351 DOI: 10.1111/mcn.13649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 02/28/2024] [Accepted: 03/11/2024] [Indexed: 04/12/2024]
Abstract
The main objective of this project was to compare in the field conditions two strategies of re-nutrition of children with moderate acute malnutrition (MAM) aged from 6 to 24 months, targeting the microbiota in comparison with a standard regimen. A three-arm, open-label, pragmatic randomised trial was conducted in four countries (Niger, CAR, Senegal and Madagascar). Children received for 12 weeks either fortified blended flour (FBF control) = arm 1, or FBF + azithromycin (oral suspension of 20 mg/kg/day daily given with a syringe) for the first 3 days at inclusion = arm 2 or mix FBF with inulin/fructo-oligosaccharides (6 g/day if age ≥12 months and 4 g if age <12 months) = arm 3. For each arm, children aged from 6 to 11 months received 100 g x 2 per day of flours and those aged from 12 to 24 months received 100 g × 3 per day of FBF. The primary endpoint was nutritional recovery, defined by reaching a weight-for-height z-score (WHZ) ≥ -1.5 within 12 weeks. Overall, 881 children were randomised (297, 290 and 294 in arm 1, arm 2 and arm 3, respectively). Three hundred and forty-four children were males (39%) and median/mean age were 14.6/14.4 months (SD = 4.9, IQR = 10.5-18.4). At inclusion, the three arms were comparable for all criteria, but differences were observed between countries. Overall, 44% (390/881) of the children recovered at week 12 from MAM, with no significant difference between the three arms (41.4%, 45.5% and 45.9%, in arm 1, arm 2 and arm 3, respectively, p = 0.47). This study did not support the true advantages of adding a prebiotic or antibiotic to flour. When using a threshold of WHZ ≥ -2 as an exploratory endpoint, significant differences were observed between the three arms, with higher success rates in arms with antibiotics or prebiotics compared to the control arm (66.9%, 66.0% and 55.2%, respectively, p = 0.005).
Collapse
Affiliation(s)
- Muriel Vray
- Emerging Diseases Epidemiology Unit, Institut de PasteurUniversité Paris‐CitéParisFrance
| | - Laura Tondeur
- Emerging Diseases Epidemiology Unit, Institut de PasteurUniversité Paris‐CitéParisFrance
| | | | | | - Alexandre Manirakiza
- Epidemiology and Clinical Research UnitInstitut Pasteur de BanguiBanguiCentral African Republic
| | | | | | - Antonio Vargas
- Nutrition and Health Unit, Action Against HungerMadridSpain
| | - André Briend
- Department of Nutrition, Exercise and Sports, Faculty of ScienceUniversity of CopenhagenFrederiksbergDenmark
- Tampere Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health TechnologyTampere University and Tampere University HospitalTampereFinland
| | - Ronan Jambou
- Direction Scientifique, Centre de Recherche Médicale et Sanitaire (CERMES)NiameyNiger
| |
Collapse
|
4
|
Baaleman DF, Wegh CAM, de Leeuw TJM, van Etten-Jamaludin FS, Vaughan EE, Schoterman MHC, Belzer C, Smidt H, Tabbers MM, Benninga MA, Koppen IJN. What are Normal Defecation Patterns in Healthy Children up to Four Years of Age? A Systematic Review and Meta-Analysis. J Pediatr 2023; 261:113559. [PMID: 37331467 DOI: 10.1016/j.jpeds.2023.113559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 06/08/2023] [Accepted: 06/12/2023] [Indexed: 06/20/2023]
Abstract
OBJECTIVE To summarize available data on defecation frequency and stool consistency of healthy children up to age 4 in order to estimate normal references values. STUDY DESIGN Systematic review including cross-sectional, observational, and interventional studies published in English, that reported on defecation frequency and/or stool consistency in healthy children 0-4 years old. RESULTS Seventy-five studies were included with 16 393 children and 40 033 measurements of defecation frequency and/or stool consistency. Based on visual inspection of defecation frequency data, a differentiation was made between two age categories: young infants (0-14 weeks old) and young children (15 weeks-4 years old). Young infants had a mean defecation frequency of 21.8 per week (95 % CI, 3.9-35.2) compared with 10.9 (CI, 5.7-16.7) in young children (P < .001). Among young infants, human milk-fed (HMF) infants had the highest mean defecation frequency per week (23.2 [CI, 8.8-38.1]), followed by formula-fed (FF) infants (13.7 [CI 5.4-23.9]), and mixed-fed (MF) infants (20.7 [CI, 7.0-30.2]). Hard stools were infrequently reported in young infants (1.5%) compared with young children (10.5%), and a reduction in the frequency of soft/watery stools was observed with higher age (27.0% in young infants compared with 6.2% in young children). HMF young infants had softer stools compared with FF young infants. CONCLUSIONS Young infants (0-14 weeks old) have softer and more frequent stools compared with young children (15 weeks-4 years old).
Collapse
Affiliation(s)
- Desiree F Baaleman
- Department of Pediatric Gastroenterology and Nutrition, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
| | - Carrie A M Wegh
- Department of Pediatric Gastroenterology and Nutrition, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Tessa J M de Leeuw
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | | | | | | | - Clara Belzer
- Department of Pediatric Gastroenterology and Nutrition, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Hauke Smidt
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Merit M Tabbers
- Department of Pediatric Gastroenterology and Nutrition, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Marc A Benninga
- Department of Pediatric Gastroenterology and Nutrition, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ilan J N Koppen
- Department of Pediatric Gastroenterology and Nutrition, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
5
|
Recent advances in oral delivery of bioactive molecules: Focus on prebiotic carbohydrates as vehicle matrices. Carbohydr Polym 2022; 298:120074. [DOI: 10.1016/j.carbpol.2022.120074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/18/2022]
|
6
|
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: 8.0] [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
|
7
|
Alam Z, Shang X, Effat K, Kanwal F, He X, Li Y, Xu C, Niu W, War AR, Zhang Y. The potential role of prebiotics, probiotics, and synbiotics in adjuvant cancer therapy especially colorectal cancer. J Food Biochem 2022; 46:e14302. [PMID: 35816322 DOI: 10.1111/jfbc.14302] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 12/13/2022]
Abstract
Cancer is a global health issue that is rising swiftly with younger people and an increased number of patients. The role of human microbiota in the pathophysiology of tumors has been paid more and more attention. Microecologics including prebiotics, probiotics, and synbiotics are among the best validated/proven resources for the application of microbiological prophylaxis and therapy. There is strong evidence that microecologics have anti-cancer activity and their potential association with cancer is significant. In this review, we will focus on the role of prebiotics, probiotics, and synbiotics in tumor suppression in maintaining the colon barrier, metabolism, immune regulation, inhibition of host tumor cell proliferation, and epidemiological-based recommendations. Besides, other signs illuminate the role of microecological agents to adjunct the cancer treatment and counter the toxic side effects of cancer drugs. In addition, we will explore their role in chemotherapy, where these probiotics can be used as an adjunct to chemotherapy, counteracting the toxic side effects of chemotherapy drugs to minimize or optimize the therapeutic effect. In the treatment of cancer, we can see the role of prebiotics, probiotics, synbiotics, and their application in cancer patients, and the effectiveness effect can be considered as a clinical benefit. PRACTICAL APPLICATIONS: A large number of studies have shown that microecologics including prebiotics, probiotics, and synbiotics play an important role in regulating intestinal microecology and contribute to the prevention and treatment of cancer, indicating that prebiotics, probiotics, and synbiotics have the potential to be used as microecological modulators in the adjuvant therapy of cancer. However, it is not clear what is the anti-tumor mechanism of these microecologics and how they antagonize the side effects of cancer chemotherapy and protect normal cells. This paper reviews the role of prebiotics, probiotics, and synbiotics in tumor suppression in maintaining the colon barrier, metabolism, immune regulation, and prevention of rapid growth of host cells, as well as their potential role in cancer chemotherapy. This review helps to better understand the relationship between prebiotics, probiotics, and synbiotics with immune regulation, intestinal microecology, metabolic regulation, and cell proliferation and provides strong evidence for their potential application as microecologics in cancer adjuvant therapy.
Collapse
Affiliation(s)
- Zahoor Alam
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Xiaoya Shang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Khansa Effat
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Freeha Kanwal
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Xiaoqin He
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yanye Li
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Chunlan Xu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Weining Niu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Abdul Rouf War
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yong Zhang
- Department of Surgical Oncology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
8
|
Keulers L, Dehghani A, Knippels L, Garssen J, Papadopoulos N, Folkerts G, Braber S, van Bergenhenegouwen J. Probiotics, prebiotics, and synbiotics to prevent or combat air pollution consequences: The gut-lung axis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 302:119066. [PMID: 35240267 DOI: 10.1016/j.envpol.2022.119066] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 05/26/2023]
Abstract
Air pollution exposure is a public health emergency, which attributes globally to an estimated seven million deaths on a yearly basis We are all exposed to air pollutants, varying from ambient air pollution hanging over cities to dust inside the home. It is a mixture of airborne particulate matter and gases that can be subdivided into three categories based on particle diameter. The smallest category called PM0.1 is the most abundant. A fraction of the particles included in this category might enter the blood stream spreading to other parts of the body. As air pollutants can enter the body via the lungs and gut, growing evidence links its exposure to gastrointestinal and respiratory impairments and diseases, like asthma, rhinitis, respiratory tract infections, Crohn's disease, ulcerative colitis, and abdominal pain. It has become evident that there exists a crosstalk between the respiratory and gastrointestinal tracts, commonly referred to as the gut-lung axis. Via microbial secretions, metabolites, immune mediators and lipid profiles, these two separate organ systems can influence each other. Well-known immunomodulators and gut health stimulators are probiotics, prebiotics, together called synbiotics. They might combat air pollution-induced systemic inflammation and oxidative stress by optimizing the microbiota composition and microbial metabolites, thereby stimulating anti-inflammatory pathways and strengthening mucosal and epithelial barriers. Although clinical studies investigating the role of probiotics, prebiotics, and synbiotics in an air pollution setting are lacking, these interventions show promising health promoting effects by affecting the gastrointestinal- and respiratory tract. This review summarizes the current data on how air pollution can affect the gut-lung axis and might impact gut and lung health. It will further elaborate on the potential role of probiotics, prebiotics and synbiotics on the gut-lung axis, and gut and lung health.
Collapse
Affiliation(s)
- Loret Keulers
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584, CG, Utrecht, the Netherlands; Danone Nutricia Research, Uppsalalaan 12, 3584, CT, Utrecht, the Netherlands.
| | - Ali Dehghani
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584, CG, Utrecht, the Netherlands
| | - Leon Knippels
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584, CG, Utrecht, the Netherlands; Danone Nutricia Research, Uppsalalaan 12, 3584, CT, Utrecht, the Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584, CG, Utrecht, the Netherlands; Danone Nutricia Research, Uppsalalaan 12, 3584, CT, Utrecht, the Netherlands
| | - Nikolaos Papadopoulos
- Centre for Paediatrics and Child Health, Institute of Human Development, University of Manchester, Oxford Road M13 9PL, Manchester, United Kingdom
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584, CG, Utrecht, the Netherlands
| | - Saskia Braber
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584, CG, Utrecht, the Netherlands
| | - Jeroen van Bergenhenegouwen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584, CG, Utrecht, the Netherlands; Danone Nutricia Research, Uppsalalaan 12, 3584, CT, Utrecht, the Netherlands
| |
Collapse
|
9
|
Goday PS, Lewis JD, Sang CJ, George DE, McGoogan KE, Safta AM, Seth A, Krekel C. Energy- and protein-enriched formula improves weight gain in infants with malnutrition due to cardiac and non-cardiac etiologies. JPEN J Parenter Enteral Nutr 2021; 46:1270-1282. [PMID: 34822187 PMCID: PMC9540590 DOI: 10.1002/jpen.2308] [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] [Indexed: 11/08/2022]
Abstract
Background We aimed to assess safety, tolerability, and improvement in weight gain with an energy‐ and protein‐enriched formula (EPEF) in infants with poor growth. Methods Infants aged 1–8 months with poor growth received EPEF for 16 weeks. Our primary objective was improvement in weight as measured by change in weight‐for‐age z‐score (WAZ) and weight gain velocity (grams per day) ≥ median for age. Secondary objectives included improvement in other anthropometric z‐scores, formula tolerance, and safety. Results Twenty‐six patients with poor growth due to congenital heart disease (n = 15), other organic causes (n = 9), and nonorganic causes (n = 2) completed the study per protocol. Mean daily energy intake was 123 ± 32 kilocalories per kilogram of body weight, with >90% of energy coming from EPEF. Weight gain velocity exceeded the median for 83% (20 of 24) and 67% (16 of 24) of infants at ≥1 time point and for the overall study period, respectively. Mean ± SD WAZ improved from −2.92 ± 1.04 at baseline to −2.01 ± 1.12 at 16 weeks (P = 0.0001). Z‐scores for weight‐for‐length and head circumference (P = 0.0001) and for length‐for‐age (P = 0.003) improved significantly at 16 weeks. Compared with baseline, stool consistency was different at 2, 4, and 16 weeks (P < 0.05). There were no significant differences in vomiting, fussiness, or daily number of stools while there was a decrease or no change in spit‐up, flatulence, crying, or gassiness. Conclusion EPEF is safe, well tolerated, and improves weight gain in infants with poor growth.
Collapse
Affiliation(s)
- Praveen S Goday
- Pediatric Gastroenterology and Nutrition, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Jeffery D Lewis
- Children's Center for Digestive Health Care, LLC, Atlanta, Georgia, USA
| | - Charlie J Sang
- Pediatric Cardiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| | - Donald E George
- Division of Pediatric Gastroenterology and Nutrition, Nemours Children's Specialty Care, Jacksonville, Florida, USA
| | - Katherine E McGoogan
- Division of Pediatric Gastroenterology and Nutrition, Nemours Children's Specialty Care, Jacksonville, Florida, USA
| | - Anca M Safta
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Anand Seth
- SK Patent Associates, LLC, Dublin, Ohio, USA
| | | |
Collapse
|
10
|
Sathitkowitchai W, Suratannon N, Keawsompong S, Weerapakorn W, Patumcharoenpol P, Nitisinprasert S, Nakphaichit M. A randomized trial to evaluate the impact of copra meal hydrolysate on gastrointestinal symptoms and gut microbiome. PeerJ 2021; 9:e12158. [PMID: 34616618 PMCID: PMC8449532 DOI: 10.7717/peerj.12158] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 08/24/2021] [Indexed: 01/04/2023] Open
Abstract
The impact of copra meal hydrolysate (CMH) on gut health was assessed by conducting a double-blinded, placebo-controlled study. Sixty healthy adult participants, aged 18–40 years were assigned to daily consume 3 g of CMH, 5 g of CMH or placebo in the form of drink powder for 21 days. Consumption of CMH at 3 g/d improved defecating conditions by reducing stool size and also relieved flatulence and bloating symptoms. Fecal samples were collected serially at the baseline before treatment, after the treatment and after a 2-week washout period. The gut microbiomes were similar among the treatment groups, with microbial community changes observed within the groups. Intake of CMH at 3 g/d led to increase microbial diversity and richness. Reduction of the ratio between Firmicutes to Bacteroidetes was observed, although it was not significantly different between the groups. The 3 g/d CMH treatment increased beneficial microbes in the group of fiber-degrading bacteria, especially human colonic Bacteroidetes, while induction of Bifidobacteriaceae was observed after the washout period. Intake of CMH led to increase lactic acid production, while 3 g/d supplement promoted the present of immunoglobulin A (IgA) in stool samples. The 3 g daily dose of CMH led to the potentially beneficial effects on gut health for healthy individuals.
Collapse
Affiliation(s)
- Witida Sathitkowitchai
- Department of Biotechnology, Faculty of Agro-Industry, Kasetsart University, Bangkok, Thailand.,Center for Advanced Studies for Agriculture and Food, Kasetsart University Institute for Advanced Studies, Kasetsart University, Bangkok, Thailand.,Microarray Research Team, National Center for Genetic Engineering and Biotechnology, Thailand Science Park, Pathum Thani, Thailand
| | - Narissara Suratannon
- Pediatric Allergy & Clinical Immunology Research Unit, Division of Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Suttipun Keawsompong
- Department of Biotechnology, Faculty of Agro-Industry, Kasetsart University, Bangkok, Thailand.,Center for Advanced Studies for Agriculture and Food, Kasetsart University Institute for Advanced Studies, Kasetsart University, Bangkok, Thailand
| | - Wanlapa Weerapakorn
- Pediatric Allergy & Clinical Immunology Research Unit, Division of Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Preecha Patumcharoenpol
- Interdisciplinary Graduate Program in Bioscience, Faculty of Science, Kasetsart University, Bangkok, Thailand
| | - Sunee Nitisinprasert
- Department of Biotechnology, Faculty of Agro-Industry, Kasetsart University, Bangkok, Thailand.,Center for Advanced Studies for Agriculture and Food, Kasetsart University Institute for Advanced Studies, Kasetsart University, Bangkok, Thailand
| | - Massalin Nakphaichit
- Department of Biotechnology, Faculty of Agro-Industry, Kasetsart University, Bangkok, Thailand.,Center for Advanced Studies for Agriculture and Food, Kasetsart University Institute for Advanced Studies, Kasetsart University, Bangkok, Thailand
| |
Collapse
|
11
|
Evaluation of an Infant Formula with Large, Milk-Phospholipid Coated Lipid Droplets on Long-Term Growth and Development of Singaporean Infants: Randomized Controlled Trial Protocol. Nutrients 2021; 13:nu13082865. [PMID: 34445029 PMCID: PMC8401090 DOI: 10.3390/nu13082865] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/16/2021] [Accepted: 08/19/2021] [Indexed: 11/24/2022] Open
Abstract
A concept infant formula (IF) was developed with physical properties of lipid droplets mimicking more closely those in human milk. This paper describes the unique design of a randomised controlled trial evaluating the impact of the concept IF on infant growth and body composition development whilst applying a cohort-like recruitment approach that fully supports breastfeeding practices of the study population. Subjects entered the study between birth and 1 months of age, and whenever parents decided to introduce formula were randomised to one of three study formulas; the concept IF comprising large lipid droplets coated by milk phospholipids and containing a specific mixture of prebiotics, a standard IF with the specific prebiotic mixture or a standard IF without the prebiotic mixture. The primary objective was to evaluate the impact of the concept IF on growth and body composition outcomes during the first year of life with a follow-up at 2, 3, 4 and 5 years of age. In addition, stool, saliva and buccal smear samples and parameters assessing safety, gastrointestinal tolerance and cognitive outcomes were collected. The applied cohort-like enrolment approach is distinctly different from standard clinical safety or efficacy studies and may provide valuable insights on trial design for the evaluation of IF while carefully considering breastfeeding practices.
Collapse
|
12
|
Kong C, Faas MM, de Vos P, Akkerman R. Impact of dietary fibers in infant formulas on gut microbiota and the intestinal immune barrier. Food Funct 2021; 11:9445-9467. [PMID: 33150902 DOI: 10.1039/d0fo01700k] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human milk (HM) is the gold standard for the nutrition of infants. An important component of HM is human milk oligosaccharides (hMOs), which play an important role in gut microbiota colonization and gut immune barrier establishment, and thereby contribute to the maturation of the immune system in early life. Guiding these processes is important as disturbances have life-long health effects and can lead to the development of allergic diseases. Unfortunately, not all infants can be exclusively fed with HM. These infants are routinely fed with infant formulas that contain hMO analogs and other non-digestible carbohydrates (NDCs) to mimic the effects of hMOs. Currently, the hMO analogs 2'-fucosyllactose (2'-FL), galacto-oligosaccharides (GOS), fructo-oligosaccharides (FOS), and pectins are added to infant formulas; however, these NDCs cannot mimic all hMO functions and therefore new NDCs and NDC mixtures need to become available for specific groups of neonates like preterm and disease-prone neonates. In this review, we discuss human data on the beneficial effects of infant formula supplements such as the specific hMO analog 2'-FL and NDCs as well as their mechanism of effects like stimulation of microbiota development, maturation of different parts of the gut immune barrier and anti-pathogenic effects. Insights into the structure-specific mechanisms by which hMOs and NDCs exert their beneficial functions might contribute to the development of new tailored NDCs and NDC mixtures. We also describe the needs for new in vitro systems that can be used for research on hMOs and NDCs. The current data suggest that "tailored infant formulas" for infants of different ages and healthy statuses are needed to ensure a healthy development of the microbiota and the gut immune system of infants.
Collapse
Affiliation(s)
- Chunli Kong
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB Groningen, The Netherlands.
| | | | | | | |
Collapse
|
13
|
Cheng L, Kong C, Wang W, Groeneveld A, Nauta A, Groves MR, Kiewiet MBG, de Vos P. The Human Milk Oligosaccharides 3-FL, Lacto-N-Neotetraose, and LDFT Attenuate Tumor Necrosis Factor-α Induced Inflammation in Fetal Intestinal Epithelial Cells In Vitro through Shedding or Interacting with Tumor Necrosis Factor Receptor 1. Mol Nutr Food Res 2021; 65:e2000425. [PMID: 33465830 PMCID: PMC8047892 DOI: 10.1002/mnfr.202000425] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 11/09/2020] [Indexed: 12/11/2022]
Abstract
SCOPE Human milk oligosaccharides (hMOs) can attenuate inflammation by modulating intestinal epithelial cells, but the mechanisms of action are not well-understood. Here, the effects of hMOs on tumor necrosis factor-α (TNF-α) induced inflammatory events in gut epithelial cells are studied. METHODS AND RESULTS The modulatory effects of 2'-fucosyllactose, 3-fucosyllactose (3-FL), 6'-sialyllactose, lacto-N-tetraose, lacto-N-neotetraose (LNnT), lactodifucotetraose (LDFT), and lacto-N-triaose (LNT2) on immature (FHs 74 Int) and adult (T84) intestinal epithelial cells with or without TNF-α are determined. Interleukin-8 (IL-8) secretion in FHs 74 Int and T84 are quantified to determine hMO induced attenuation of inflammatory events by ELISA. 3-FL, LNnT, and LDFT significantly attenuate TNF-α induced inflammation in FHs 74 Int, while LNT2 induces IL-8 secretion in T84. In addition, microscale thermophoresis assays and ELISA are used to study the possible mechanisms of interaction between effective hMOs and tumor necrosis factor receptor 1 (TNFR1). 3-FL, LNnT, and LDFT exert TNFR1 ectodomain shedding while LNnT also shows binding affinity to TNFR1 with a Kd of 900 ± 660 nM. CONCLUSION The findings indicate that specific hMO types attenuate TNF-α induced inflammation in fetal gut epithelial cells through TNFR1 in a hMO structure-dependent fashion suggest possibilities to apply hMOs in management of TNF-α dependent diseases.
Collapse
Affiliation(s)
- Lianghui Cheng
- Immunoendocrinology, Division of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen, University Medical Center GroningenHanzeplein 1, RBGroningen9700The Netherlands
| | - Chunli Kong
- Immunoendocrinology, Division of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen, University Medical Center GroningenHanzeplein 1, RBGroningen9700The Netherlands
| | - Wenjia Wang
- Drug Design XB20, Groningen Research Institute of PharmacyUniversity of GroningenADGroningen9700The Netherlands
| | - Andre Groeneveld
- FrieslandCampinaStationsplein 4, LEAmersfoort3818The Netherlands
| | - Arjen Nauta
- FrieslandCampinaStationsplein 4, LEAmersfoort3818The Netherlands
| | - Matthew R. Groves
- Drug Design XB20, Groningen Research Institute of PharmacyUniversity of GroningenADGroningen9700The Netherlands
| | - Mensiena B. G. Kiewiet
- Immunoendocrinology, Division of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen, University Medical Center GroningenHanzeplein 1, RBGroningen9700The Netherlands
| | - Paul de Vos
- Immunoendocrinology, Division of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen, University Medical Center GroningenHanzeplein 1, RBGroningen9700The Netherlands
| |
Collapse
|
14
|
Younes M, Aquilina G, Castle L, Engel K, Fowler P, Frutos Fernandez MJ, Fürst P, Gürtler R, Husøy T, Manco M, Mennes W, Moldeus P, Passamonti S, Shah R, Waalkens‐Berendsen I, Wölfle D, Wright M, Dusemund B, Mortensen A, Turck D, Barmaz S, Tard A, Vianello G, Gundert‐Remy U. Opinion on the re-evaluation of pectin (E 440i) and amidated pectin (E 440ii) as food additives in foods for infants below 16 weeks of age and follow-up of their re-evaluation as food additives for uses in foods for all population groups. EFSA J 2021; 19:e06387. [PMID: 33537069 PMCID: PMC7845505 DOI: 10.2903/j.efsa.2021.6387] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Pectin (E 440i) and amidated pectin (E 440ii) were re-evaluated in 2017 by the former EFSA Panel on Food Additives and Nutrient sources added to Food (ANS). As a follow-up to this assessment, the Panel on Food Additives and Flavourings (FAF) was requested to assess the safety of pectins (E 440i,ii) for their uses as food additives in food for infants below 16 weeks of age. In addition, the FAF Panel was requested to address the issues already identified during the re-evaluation of the same food additive. The process involved the publication of a call for data to allow the interested business operators to provide the requested information to complete the risk assessment. Based on the information submitted in response to the call for data, the FAF Panel considered it feasible to amend the current specifications, in particular for the toxic elements arsenic, lead, cadmium, mercury and for sulfur dioxide and to introduce new specifications for aluminium and microbiological criteria. Studies on neonatal piglets, clinical studies and post-marketing data were made available during the call for data. Due to the low internal validity of the clinical studies, the Panel concluded that a reference point could not be derived from them, but the results of the adequate piglet study could serve to derive a reference point. When calculating the margin of safety for pectins exposure, this was below 1 for some scenarios. At the maximum permitted levels (MPLs), an internal methanol dose would be produced that could lead to adverse health effects in infants below 16 weeks of age. The FAF Panel recommended a reduction of the MPL of pectin (E 440i) and amidated pectin (E 440ii) in food categories 13.1.5.1 and 13.1.5.2, in order to reduce the exposure to both the additives themselves and to methanol.
Collapse
|
15
|
Dietary pectin at 0.2% in milk replacer did not inhibit growth, feed intake, or nutrient digestibility in a 3-week neonatal pig study. Regul Toxicol Pharmacol 2020; 114:104669. [DOI: 10.1016/j.yrtph.2020.104669] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 04/06/2020] [Accepted: 04/24/2020] [Indexed: 01/30/2023]
|
16
|
Sanchez-Siles LM, Bernal MJ, Gil D, Bodenstab S, Haro-Vicente JF, Klerks M, Plaza-Diaz J, Gil Á. Are Sugar-Reduced and Whole Grain Infant Cereals Sensorially Accepted at Weaning? A Randomized Controlled Cross-Over Trial. Nutrients 2020; 12:1883. [PMID: 32599738 PMCID: PMC7353261 DOI: 10.3390/nu12061883] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/19/2020] [Accepted: 06/22/2020] [Indexed: 01/10/2023] Open
Abstract
The way infants are fed during the complementary period can have a significant impact on infants' health and development. Infant cereals play an important role in complementary feeding in many countries. In spite of well documented benefits of a low sugar and high whole grain diet, commercial infant cereals are often refined and contain a high amount of sugars. The aim of the present study was to compare the sensory acceptability, gastrointestinal tolerance and bowel habits of two commercially available infant cereals in Spain with varying sugar and whole grain contents in infants at weaning. Forty-six healthy infants (mean age = 5.2 ± 0.4 months) received one of the two infant cereals containing either 0% whole grain flour and a high sugar content produced by starch hydrolysis (24 g/100 g) (Cereal A) or 50% whole grain flour and a medium-sugar content produced by hydrolysis (12 g/100 g) (Cereal B) in a randomized, triple blind, cross-over controlled trial. Both types of infant cereals were consumed for seven weeks. The cross-over was carried out after seven weeks. Sensory acceptability, anthropometry, gastrointestinal tolerance and adverse events were measured, and results evaluated using a linear regression model. No significant differences were observed between groups in any of the main variables analyzed. Importantly, the long-term health implications of our findings represent a wake-up call for the food industry to reduce or even eliminate simple sugars in infant cereals and for regulatory bodies and professional organizations to recommend whole grain infant cereals.
Collapse
Affiliation(s)
- Luis Manuel Sanchez-Siles
- Research and Nutrition Lab, Hero Group, 30820 Murcia, Spain; (M.J.B.); (J.F.H.-V.); (M.K.)
- Institute for Research and Nutrition, Hero Group, 5600 Lenzburg, Switzerland;
| | - Maria Jose Bernal
- Research and Nutrition Lab, Hero Group, 30820 Murcia, Spain; (M.J.B.); (J.F.H.-V.); (M.K.)
| | - David Gil
- Pediatric Gastroenterology, Hepatology and Nutrition Unit, Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain;
| | - Stefan Bodenstab
- Institute for Research and Nutrition, Hero Group, 5600 Lenzburg, Switzerland;
| | | | - Michelle Klerks
- Research and Nutrition Lab, Hero Group, 30820 Murcia, Spain; (M.J.B.); (J.F.H.-V.); (M.K.)
| | - Julio Plaza-Diaz
- Institute of Nutrition and Food Technology “José Mataix”, Center of Biomedical Research, University of Granada, Avda. del Conocimiento s/n., 18016 Armilla, Granada, Spain; (J.P.-D.); (Á.G.)
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- ibs.GRANADA, Instituto de Investigación Biosanitaria, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
| | - Ángel Gil
- Institute of Nutrition and Food Technology “José Mataix”, Center of Biomedical Research, University of Granada, Avda. del Conocimiento s/n., 18016 Armilla, Granada, Spain; (J.P.-D.); (Á.G.)
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- ibs.GRANADA, Instituto de Investigación Biosanitaria, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
- CIBEROBN (CIBER Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
17
|
Cheng L, Kong C, Walvoort MTC, Faas MM, de Vos P. Human Milk Oligosaccharides Differently Modulate Goblet Cells Under Homeostatic, Proinflammatory Conditions and ER Stress. Mol Nutr Food Res 2020; 64:e1900976. [PMID: 31800974 PMCID: PMC7079026 DOI: 10.1002/mnfr.201900976] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 11/21/2019] [Indexed: 12/14/2022]
Abstract
SCOPE Human milk oligosaccharides (hMOs) have beneficial effects on intestinal barrier function, but the mechanisms of action are not well understood. Here, the effects of hMOs on goblet cells, which indicate that some hMOs may enhance mucus barrier function through direct modulation of goblet cell function, are studied. METHODS AND RESULTS The modulatory effects of 2'-fucosyllactose (2'-FL), 3-fucosyllactose (3-FL), lacto-N-triaose II (LNT2), and galacto-oligosaccharides (GOS) on the expression of goblet cell secretory related genes MUC2, TFF3, and RETNLB, and the Golgi-sulfotransferase genes CHST5 and GAL3ST2 of LS174T are determined by real-time quantitative RT-PCR. 3-FL, LNT2, and GOS-modulated LS174T gene expression profiles in a dose- and time-dependent manner. In addition, the upregulation of MUC2 is confirmed by immunofluorescence staining. Effects of 2'-FL, 3-FL, LNT2, and GOS on gene transcription of LS174T are also assessed during exposure to TNF-α, IL-13, or tunicamycin. During TNF-α challenge, 3-FL and LNT2 enhance MUC2 and TFF3 gene expression. After IL-13 exposure, 2'-FL, 3-FL, and LNT2 all show upregulating effects on MUC2; 3-FL and LNT2 also enhance TFF3 expression. LNT2 significantly reverses Tm-induced downregulation of TFF3, RETNLB, and CHST5. CONCLUSION The findings indicate that hMOs may enhance mucus barrier function through direct modulation of intestinal goblet cells. Effects are structure- and stressor-dependent.
Collapse
Affiliation(s)
- Lianghui Cheng
- Immunoendocrinology, Division of Medical BiologyDepartment of Pathology and Medical BiologyUniversity Medical Center GroningenUniversity of GroningenHanzeplein 19700 RBGroningenThe Netherlands
| | - Chunli Kong
- Immunoendocrinology, Division of Medical BiologyDepartment of Pathology and Medical BiologyUniversity Medical Center GroningenUniversity of GroningenHanzeplein 19700 RBGroningenThe Netherlands
| | - Marthe T. C. Walvoort
- Stratingh Institute for Chemistry, Faculty of Science and EngineeringUniversity of Groningen9700 RBGroningenThe Netherlands
| | - Marijke M. Faas
- Immunoendocrinology, Division of Medical BiologyDepartment of Pathology and Medical BiologyUniversity Medical Center GroningenUniversity of GroningenHanzeplein 19700 RBGroningenThe Netherlands
| | - Paul de Vos
- Immunoendocrinology, Division of Medical BiologyDepartment of Pathology and Medical BiologyUniversity Medical Center GroningenUniversity of GroningenHanzeplein 19700 RBGroningenThe Netherlands
| |
Collapse
|
18
|
Kong C, Elderman M, Cheng L, de Haan BJ, Nauta A, de Vos P. Modulation of Intestinal Epithelial Glycocalyx Development by Human Milk Oligosaccharides and Non-Digestible Carbohydrates. Mol Nutr Food Res 2019; 63:e1900303. [PMID: 31140746 PMCID: PMC6771538 DOI: 10.1002/mnfr.201900303] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 03/26/2019] [Indexed: 12/11/2022]
Abstract
SCOPE The epithelial glycocalyx development is of great importance for microbial colonization. Human milk oligosaccharides (hMOs) and non-digestible carbohydrates (NDCs) may modulate glycocalyx development. METHODS AND RESULTS The effects of hMOs and NDCs on human gut epithelial cells (Caco2) are investigated by quantifying thickness and area coverage of adsorbed albumin, heparan sulfate (HS), and hyaluronic acid (HA) in the glycocalyx. Effects of hMOs (2'-FL and 3-FL) and NDCs [inulins with degrees of polymerization (DP) (DP3-DP10, DP10-DP60, DP30-DP60) and pectins with degrees of methylation (DM) (DM7, DM55, DM69)] are tested using immunofluorescence staining at 1 and 5 days stimulation. HMOs show a significant enhancing effect on glycocalyx development but effects are structure-dependent. 3-FL induces a stronger albumin adsorption and increases HS and HA stronger than 2'-FL. The DP3-DP10, DP30-60 inulins also increase glycocalyx development in a structure-dependent manner as DP3-DP10 selectively increases HS, while DP30-DP60 specifically increases HA. Pectins have less effects, and only increase albumin adsorption. CONCLUSION Here, it is shown that 2'-FL and 3-FL and inulins stimulate glycocalyx development in a structure-dependent fashion. This may contribute to formulation of effective hMO and NDC formulations in infant formulas to support microbial colonization and gut barrier function.
Collapse
Affiliation(s)
- Chunli Kong
- Immunoendocrinology GroupDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity Medical Center Groningen, University of GroningenHanzeplein 19700 RBGroningenThe Netherlands
| | - Marlies Elderman
- Immunoendocrinology GroupDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity Medical Center Groningen, University of GroningenHanzeplein 19700 RBGroningenThe Netherlands
| | - Lianghui Cheng
- Immunoendocrinology GroupDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity Medical Center Groningen, University of GroningenHanzeplein 19700 RBGroningenThe Netherlands
| | - Bart J. de Haan
- Immunoendocrinology GroupDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity Medical Center Groningen, University of GroningenHanzeplein 19700 RBGroningenThe Netherlands
| | - Arjen Nauta
- FrieslandCampinaStationsplein 43818 LEAmersfoortThe Netherlands
| | - Paul de Vos
- Immunoendocrinology GroupDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity Medical Center Groningen, University of GroningenHanzeplein 19700 RBGroningenThe Netherlands
| |
Collapse
|
19
|
Benninga MA, Vandenplas Y. The Magnesium-Rich Formula for Functional Constipation in Infants: a Randomized Comparator-Controlled Study. Pediatr Gastroenterol Hepatol Nutr 2019; 22:270-281. [PMID: 31110960 PMCID: PMC6506425 DOI: 10.5223/pghn.2019.22.3.270] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 11/12/2018] [Accepted: 11/24/2018] [Indexed: 12/02/2022] Open
Abstract
PURPOSE To compare the effectiveness of the magnesium (Mg)-enriched formula vs. control formula in constipated infants. METHODS An open-label, interventional, and the comparator-controlled study was conducted to evaluate the effectiveness of the Mg-enriched formula in formula-fed infants ≤6 months old presenting with functional constipation according to modified Rome IV criteria. Infants were randomized 1:1 to intervention or control formula for 30 days. Parents recorded stool consistency (hard, normal, or watery) and frequency on days 1-7 and 23-29. Physicians recorded patient baseline characteristics and performed the clinical examination at the time of three patient visits (baseline, day 8, and 30). RESULTS Of the 286 recruited infants, 143 received the Mg-rich formula and 142 received the control formula. After 7 days, significantly more infants had stools with normal consistency with the Mg-rich formula compared to the infants fed with the control formula (81.8% vs. 41.1%; p<0.001). The number of infants passing one or more stools per day was increased at day 7 in the Mg-rich formula group (86.7% vs. 68.2%; p<0.001). At days 7 and 29, >25% of infants responded completely to the Mg-rich formula compared to <5% of infants fed with the control formula (p<0.001). Parents of infants in the Mg-rich formula group were very satisfied with the treatment (80.8% vs. 10.2%), with the majority willing to continue treatment after 30 days (97.9% vs. 52.6%; p<0.001). CONCLUSION The Mg-rich formula significantly improved stool consistency and frequency compared to the control formula in constipated infants.
Collapse
Affiliation(s)
- Marc A Benninga
- Department of Pediatric Gastroenterology, Emma Children's Hospital, Academic Medical Center, Amsterdam, Netherlands
| | | | - Yvan Vandenplas
- KidZ Health Castle, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
20
|
Vray M, Hedible BG, Adam P, Tondeur L, Manirazika A, Randremanana R, Mainassara H, Briend A, Artaud C, von Platen C, Altmann M, Jambou R. A multicenter, randomized controlled comparison of three renutrition strategies for the management of moderate acute malnutrition among children aged from 6 to 24 months (the MALINEA project). Trials 2018; 19:666. [PMID: 30514364 PMCID: PMC6278112 DOI: 10.1186/s13063-018-3027-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 10/29/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The aim of this open-label, randomized controlled trial conducted in four African countries (Madagascar, Niger, Central African Republic, and Senegal) is to compare three strategies of renutrition for moderate acute malnutrition (MAM) in children based on modulation of the gut microbiota with enriched flours alone, enriched flours with prebiotics or enriched flours coupled with antibiotic treatment. METHODS To be included, children aged between 6 months and 2 years are preselected based on mid-upper-arm circumference (MUAC) and are included based on a weight-for-height Z-score (WHZ) between - 3 and - 2 standard deviations (SD). As per current protocols, children receive renutrition treatment for 12 weeks and are assessed weekly to determine improvement. The primary endpoint is recovery, defined by a WHZ ≥ - 1.5 SD after 12 weeks of treatment. Data collected include clinical and socioeconomic characteristics, side effects, compliance and tolerance to interventions. Metagenomic analysis of gut microbiota is conducted at inclusion, 3 months, and 6 months. The cognitive development of children is evaluated in Senegal using only the Developmental Milestones Checklist II (DMC II) questionnaire at inclusion and at 3, 6, and 9 months. The data will be correlated with renutrition efficacy and metagenomic data. DISCUSSION This study will provide new insights for the treatment of MAM, as well as original data on the modulation of gut microbiota during the renutrition process to support (or not) the microbiota hypothesis of malnutrition. TRIAL REGISTRATION ClinicalTrials.gov, ID: NCT03474276 Last update 28 May 2018.
Collapse
Affiliation(s)
- Muriel Vray
- Unité d’Epidémiologie des Maladies Infectieuses, Institut Pasteur Dakar, Dakar, Senegal
- Unité des Epidémies et des Maladies Emergentes, Institut Pasteur, 25 Rue du Dr. Roux, 75015 Paris, France
| | - Boris G. Hedible
- Unité d’Epidémiologie des Maladies Infectieuses, Institut Pasteur Dakar, Dakar, Senegal
| | - Pierrick Adam
- Unité des Epidémies et des Maladies Emergentes, Institut Pasteur, 25 Rue du Dr. Roux, 75015 Paris, France
| | - Laura Tondeur
- Unité des Epidémies et des Maladies Emergentes, Institut Pasteur, 25 Rue du Dr. Roux, 75015 Paris, France
| | - Alexandre Manirazika
- Unité d’Epidémiologie Institut Pasteur de Bangui, Bangui, Central African Republic
| | - Rindra Randremanana
- Unité d’Epidémiologie, Institut Pasteur de Madagascar, BP1274, 101 Antananarivo, Madagascar
| | | | - André Briend
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Rolighedsvej 30, DK-1958 Frederiksberg, Denmark
- Tampere Centre for Child Health Research, University of Tampere, Lääkärinkatu 1, 33014 Tampere, Finland
| | - Cecile Artaud
- Centre de recherche Transactionnel, Institut Pasteur, 28 Rue du Dr. Roux, 75015 Paris, France
| | - Cassandre von Platen
- Centre de recherche Transactionnel, Institut Pasteur, 28 Rue du Dr. Roux, 75015 Paris, France
| | - Mathias Altmann
- Action Contre la Faim, 14/16 Boulevard Douaumont – CS 80060, PARIS CEDEX 17, 75854 Paris, France
| | - Ronan Jambou
- Department of Parasites and Vector Insects, Institut Pasteur, 28 Rue du Dr. Roux, 75015 Paris, France
| |
Collapse
|
21
|
Cuello-Garcia C, Fiocchi A, Pawankar R, Yepes-Nuñez JJ, Morgano GP, Zhang Y, Agarwal A, Gandhi S, Terracciano L, Schünemann HJ, Brozek JL. Prebiotics for the prevention of allergies: A systematic review and meta-analysis of randomized controlled trials. Clin Exp Allergy 2018; 47:1468-1477. [PMID: 29035013 DOI: 10.1111/cea.13042] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 09/07/2017] [Accepted: 09/12/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND Prevalence of allergic diseases in infants is approximately 10% reaching 20 to 30% in those with an allergic first-degree relative. Prebiotics are selectively fermented food ingredients that allow specific changes in composition/activity of the gastrointestinal microflora. They modulate immune responses, and their supplementation has been proposed as an intervention to prevent allergies. OBJECTIVE To assess in pregnant women, breastfeeding mothers, and infants (populations) the effect of supplementing prebiotics (intervention) versus no prebiotics (comparison) on the development of allergic diseases and to inform the World Allergy Organization guidelines. METHODS We performed a systematic review of studies assessing the effects of prebiotic supplementation with an intention to prevent the development of allergies. RESULTS Of 446 unique records published until November 2016 in Cochrane, MEDLINE, and EMBASE, 22 studies fulfilled a priori specified criteria. We did not find any studies of prebiotics given to pregnant women or breastfeeding mothers. Prebiotic supplementation in infants, compared to placebo, had the following effects: risk of developing eczema (RR: 0.68, 95% CI: 0.40 to 1.15), wheezing/asthma (RR, 0.37; 95% CI: 0.17 to 0.80), and food allergy (RR: 0.28, 95% CI: 0.08 to 1.00). There was no evidence of an increased risk of any adverse effects (RR: 1.01, 95% CI: 0.92 to 1.10). Prebiotic supplementation had little influence growth rate (MD: 0.92 g per day faster with prebiotics, 95% CI: 0 to 1.84) and the final infant weight (MD: 0.10 kg higher with prebiotics, 95% CI: -0.09 to 0.29). The certainty of these estimates is very low due to risk of bias and imprecision of the results. CONCLUSIONS Currently available evidence on prebiotic supplementation to reduce the risk of developing allergies is very uncertain.
Collapse
Affiliation(s)
- C Cuello-Garcia
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - A Fiocchi
- Pediatric Hospital Bambino Gesù, Rome, Italy
| | - R Pawankar
- Department of Pediatrics, Nippon Medical School, Tokyo, Japan
| | - J J Yepes-Nuñez
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada.,University of Antioquia School of Medicine, Medellín, Colombia
| | - G P Morgano
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - Y Zhang
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - A Agarwal
- Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - S Gandhi
- Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - L Terracciano
- Department of Child and Maternal Medicine, University of Milan Medical School at the Melloni Hospital, Milan, Italy
| | - H J Schünemann
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - J L Brozek
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
22
|
Infant formulae supplemented with prebiotics: Are they better than unsupplemented formulae? An updated systematic review. Br J Nutr 2018; 119:810-825. [DOI: 10.1017/s0007114518000120] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
AbstractIn 2011, the Committee on Nutrition of the European Society for Paediatric Gastroenterology, Hepatology and Nutrition systematically reviewed published evidence related to the safety and health effects of the administration of formulae supplemented with pro- and/or prebiotics compared with unsupplemented formulae. We updated evidence on the effects of the administration of prebiotic-supplemented infant formulae (IF) compared with unsupplemented IF. Five databases were searched up to March 2017 for randomised controlled trials. In all, forty-one publications were identified, including twenty-five new publications. The administration of currently evaluated prebiotic-supplemented formulae to healthy infants does not raise safety concerns with regard to growth and adverse effects. Some favourable clinical effects are possible, primarily stool softening, which may be beneficial in some infants. Currently, there is no existing robust evidence to recommend the routine use of prebiotic-supplemented formulae. The latter conclusion may reflect the small amount of data on specific prebiotics and outcomes, rather than a genuine lack of an effect. The efficacy and safety should be considered for each prebiotic(s)-supplemented formula.
Collapse
|
23
|
Nadia L, Anna O, Camilla M, Pasqua P, Laura M, Chiara Cristiana C, Maria Lorella G, Paola R, Fabio M. Clinical evaluation of two different protein content formulas fed to full-term healthy infants: a randomized controlled trial. BMC Pediatr 2018; 18:59. [PMID: 29439736 PMCID: PMC5812222 DOI: 10.1186/s12887-018-1046-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 02/05/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND A high early protein intake is associated with rapid postnatal weight gain and altered body composition. We aimed to evaluate the safety of a low-protein formula in healthy full-term infants. METHODS A randomized controlled trial was conducted. A total of 118 infants were randomized to receive two different protein content formulas (formula A or formula B (protein content: 1.2 vs. 1.7 g/100 mL, respectively)) for the first 4 months of life. Anthropometry and body composition by air displacement plethysmography were assessed at enrolment and at two and 4 months. The reference group comprised 50 healthy, exclusively breastfed, full-term infants. RESULTS Weight gain (g/day) throughout the study was similar between the formula groups (32.5 ± 6.1 vs. 32.8 ± 6.8) and in the reference group (30.4 ± 5.4). The formula groups showed similar body composition but a different fat-free mass content from breastfed infants at two and 4 months. However, the formula A group showed a fat-free mass increase more similar to that of the breastfed infants. The occurrence of gastrointestinal symptoms or adverse events was similar between the formula groups. CONCLUSIONS Feeding a low-protein content formula appears to be safe and to promote adequate growth, although determination of the long-term effect on body composition requires further study. TRIAL REGISTRATION The present study was retrospectively registered in ClinicalTrials.gov (trial number: NCT03035721 on January 18, 2017).
Collapse
Affiliation(s)
- Liotto Nadia
- Fondazione I.R.C.C.S. Ca Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit Department of Clinical Science and Community Health, University of Milan, Milan, Italy
- Centro di Nutrizione a Partenza neonatale, Clinica Mangiagalli, Via Della Commenda, 12, 20122 Milan, Italy
| | - Orsi Anna
- Fondazione I.R.C.C.S. Ca Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit Department of Clinical Science and Community Health, University of Milan, Milan, Italy
| | - Menis Camilla
- Fondazione I.R.C.C.S. Ca Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit Department of Clinical Science and Community Health, University of Milan, Milan, Italy
| | - Piemontese Pasqua
- Fondazione I.R.C.C.S. Ca Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit Department of Clinical Science and Community Health, University of Milan, Milan, Italy
| | - Morlacchi Laura
- Fondazione I.R.C.C.S. Ca Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit Department of Clinical Science and Community Health, University of Milan, Milan, Italy
| | - Condello Chiara Cristiana
- Fondazione I.R.C.C.S. Ca Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit Department of Clinical Science and Community Health, University of Milan, Milan, Italy
| | - Giannì Maria Lorella
- Fondazione I.R.C.C.S. Ca Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit Department of Clinical Science and Community Health, University of Milan, Milan, Italy
| | - Roggero Paola
- Fondazione I.R.C.C.S. Ca Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit Department of Clinical Science and Community Health, University of Milan, Milan, Italy
| | - Mosca Fabio
- Fondazione I.R.C.C.S. Ca Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit Department of Clinical Science and Community Health, University of Milan, Milan, Italy
| |
Collapse
|
24
|
Mortensen A, Aguilar F, Crebelli R, Di Domenico A, Dusemund B, Frutos MJ, Galtier P, Gott D, Gundert-Remy U, Lambré C, Leblanc JC, Lindtner O, Moldeus P, Mosesso P, Oskarsson A, Parent-Massin D, Stankovic I, Waalkens-Berendsen I, Wright M, Younes M, Tobback P, Ioannidou S, Tasiopoulou S, Woutersen RA. Re-evaluation of pectin (E 440i) and amidated pectin (E 440ii) as food additives. EFSA J 2017; 15:e04866. [PMID: 32625540 PMCID: PMC7010145 DOI: 10.2903/j.efsa.2017.4866] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Following a request from the European Commission, the EFSA Panel on Food Additives and Nutrient sources added to Food (ANS) was asked to deliver a scientific opinion on the re-evaluation of pectin (E 440i) and amidated pectin (E 440ii) as food additives. An acceptable daily intake (ADI) 'not specified' was allocated by the Scientific Committee for Food (SCF) for E 440i and E 440ii. Pectin and amidated pectin would not be absorbed intact, but extensively fermented by intestinal microbiota in animals and humans; products formed from pectins in the gastrointestinal tract are similar to manufactured pectin-derived acidic oligosaccharides (pAOS). There is no indication of genotoxicity for pectin and amidated pectin, although the available data were limited. No adverse effects were reported in a chronic toxicity study in rats at levels up to 5,000 mg pectin/kg bw per day, the highest dose tested. No treatment-related effects were observed in a dietary one-generation reproductive toxicity study with pAOS in rats at up to 6,200 mg/kg body weight (bw) per day, the highest dose tested. The Panel did not consider E 440i and E 440ii as having allergenic potential. A dose of 36 g/day (equivalent to 515 mg/kg bw per day) for 6 weeks in humans was without adverse effects. Exposure to pectins from their use as food additives ranged up to 442 mg/kg bw per day for toddlers at the 95th percentile (brand-loyal scenario). The Panel concluded that there is no safety concern for the use of pectin (E 440i) and amidated pectin (E 440ii) as food additives for the general population and that there is no need for a numerical ADI.
Collapse
|
25
|
Constable A, Mahadevan B, Pressman P, Garthoff JA, Meunier L, Schrenk D, Speijers G, O’Sullivan A, Hayes AW. An integrated approach to the safety assessment of food additives in early life. TOXICOLOGY RESEARCH AND APPLICATION 2017. [DOI: 10.1177/2397847317707370] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
During the development of international standards by the Codex Alimentarius Commission, infant foods and their constituent ingredients are subject to rigorous risk analysis and are strictly regulated by many authorities. Various jurisdictions have approved only a limited number of additives specifically with regard to infant foods to fulfill specific technical requirements of quality. As part of the approval process, a rigorous safety assessment is essential to confirm that the use of additives does not pose any health risk for the consumer. An acceptable daily intake (ADI) may be derived from the toxicological databases. However, the ADI may not be applicable to infants because of the possible developmental sensitivities and potentially high exposure scenarios, leading to possible lower margins of safety than would often be determined for adult populations. There is interest in defining better food safety assessment approaches for pre-weaned infants aged less than 12–16 weeks. To confirm safe use in infants, we reviewed the suitability of the existing safety databases of six additives with historical uses in infant nutrition products. To determine further toxicity testing strategies, it is necessary to understand whether the chemical used in the additives is identical to endogenous physiological metabolites and/or whether immature organs of infants are targets of toxicity. Combined with an in-depth review of the existing relevant toxicological and nutritional studies, this integrated approach will facilitate decision-making. We propose a decision tree as a tool within this approach to help guide appropriate data requirements and identify data gaps. In cases of reasonable uncertainty, studies of targeted juvenile should be considered to investigate the safe use levels in food products.
Collapse
Affiliation(s)
| | | | - Peter Pressman
- Division of Medicine, Public Health & Nutrition, The Daedalus Foundation, Alexandria, VA, USA
| | | | - Leo Meunier
- Danone Food Safety Center, Uppsalalaan, Utrecht, The Netherlands
| | - Dieter Schrenk
- Food Chemistry and Toxicology, University of Kaiserslautern, Kaiserslautern, Germany
| | - Gerrit Speijers
- General Health Effects Toxicology Safety Food (GETS), Nieuwegein, The Netherlands
| | - Aaron O’Sullivan
- Danone Trading Medical BV, Schiphol Boulevard, Schiphol Airport, The Netherlands
| | - A Wallace Hayes
- Harvard University, Boston, MA, USA and Michigan State University, East Lansing, MI, USA
| |
Collapse
|
26
|
Carter GM, Esmaeili A, Shah H, Indyk D, Johnson M, Andreae M, Sacks HS. Probiotics in Human Immunodeficiency Virus Infection: A Systematic Review and Evidence Synthesis of Benefits and Risks. Open Forum Infect Dis 2016; 3:ofw164. [PMID: 27747250 PMCID: PMC5063545 DOI: 10.1093/ofid/ofw164] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 07/25/2016] [Indexed: 12/11/2022] Open
Abstract
The risk of sepsis is low. The best probiotic to optimize outcomes has not yet been identified. Potential benefit for CD4 count, recurrence or management of bacterial vaginosis and diarrhea. Uncertain effect on translocation, BV treatment. People living with human immunodeficiency virus frequently use dietary supplements, including probiotics, but concern exists about ingesting live organisms. We performed a systematic review of the benefits of probiotics and a meta-analysis of sepsis risk. We undertook a protocol-driven, comprehensive review to identify all relevant studies, assess their quality, and summarize the evidence. Of 2068 references, 27 were analyzed. The data suggest possible benefits for CD4 count, recurrence or management of bacterial vaginosis, and diarrhea management. We examined randomized, controlled studies explicitly assessing sepsis in any patient population, and we found zero cases of supplement-associated bacteremia or fungemia in 39 randomized controlled trials comprising 9402 subjects. The estimated number needed to harm is 7369 in Bayesian approach (95% credible interval: 1689, ∞), which should reassure clinicians. No or mild adverse effects were reported. Longer duration studies investigating different individual and mixed strains for plausible indications are needed to establish best practices.
Collapse
Affiliation(s)
| | | | | | - Debbie Indyk
- Department of Preventive Medicine , Icahn School of Medicine at Mount Sinai
| | | | - Michael Andreae
- Department of Anesthesiology , Albert Einstein College of Medicine , Bronx, New York
| | - Henry S Sacks
- Department of Preventive Medicine , Icahn School of Medicine at Mount Sinai
| |
Collapse
|
27
|
Cuello-Garcia CA, Fiocchi A, Pawankar R, Yepes-Nuñez JJ, Morgano GP, Zhang Y, Ahn K, Al-Hammadi S, Agarwal A, Gandhi S, Beyer K, Burks W, Canonica GW, Ebisawa M, Kamenwa R, Lee BW, Li H, Prescott S, Riva JJ, Rosenwasser L, Sampson H, Spigler M, Terracciano L, Vereda A, Waserman S, Schünemann HJ, Brożek JL. World Allergy Organization-McMaster University Guidelines for Allergic Disease Prevention (GLAD-P): Prebiotics. World Allergy Organ J 2016; 9:10. [PMID: 26962387 PMCID: PMC4772464 DOI: 10.1186/s40413-016-0102-7] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 02/05/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The prevalence of allergic diseases in infants, whose parents and siblings do not have allergy, is approximately 10 % and reaches 20-30 % in those with an allergic first-degree relative. Intestinal microbiota may modulate immunologic and inflammatory systemic responses and, thus, influence development of sensitization and allergy. Prebiotics - non-digestible oligosaccharides that stimulate growth of probiotic bacteria - have been reported to modulate immune responses and their supplementation has been proposed as a preventive intervention. OBJECTIVE The World Allergy Organization (WAO) convened a guideline panel to develop evidence-based recommendations about the use of prebiotics in the prevention of allergy. METHODS The WAO guideline panel identified the most relevant clinical questions about the use of prebiotics for the prevention of allergy. We performed a systematic review of randomized controlled trials of prebiotics, and reviewed the evidence about patient values and preferences, and resource requirements (up to January 2015, with an update on July 29, 2015). We followed the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach to develop recommendations. RESULTS Based on GRADE evidence to decision frameworks, the WAO guideline panel suggests using prebiotic supplementation in not-exclusively breastfed infants and not using prebiotic supplementation in exclusively breastfed infants. Both recommendations are conditional and based on very low certainty of the evidence. We found no experimental or observational study of prebiotic supplementation in pregnant women or in breastfeeding mothers. Thus, the WAO guideline panel chose not to provide a recommendation about prebiotic supplementation in pregnancy or during breastfeeding, at this time. CONCLUSIONS WAO recommendations about prebiotic supplementation for the prevention of allergy are intended to support parents, clinicians and other health care professionals in their decisions whether or not to use prebiotics for the purpose of preventing allergies in healthy, term infants.
Collapse
Affiliation(s)
- Carlos A. Cuello-Garcia
- />Department of Clinical Epidemiology & Biostatistics, McMaster University, Hamilton, ON Canada
- />Tecnologico de Monterrey School of Medicine, Monterrey, Mexico
| | | | - Ruby Pawankar
- />Department of Pediatrics, Nippon Medical School, Tokyo, Japan
| | - Juan José Yepes-Nuñez
- />Department of Clinical Epidemiology & Biostatistics, McMaster University, Hamilton, ON Canada
- />University of Antioquia, School of Medicine, Medellín, Colombia
| | - Gian Paolo Morgano
- />Department of Clinical Epidemiology & Biostatistics, McMaster University, Hamilton, ON Canada
| | - Yuan Zhang
- />Department of Clinical Epidemiology & Biostatistics, McMaster University, Hamilton, ON Canada
| | - Kangmo Ahn
- />Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Suleiman Al-Hammadi
- />Department of Pediatrics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Arnav Agarwal
- />Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | - Shreyas Gandhi
- />Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | | | - Wesley Burks
- />Department of Pediatrics, University of North Carolina, Chapel Hill, NC USA
| | | | - Motohiro Ebisawa
- />Department of Allergy, Clinical Research Center for Allergology and Rheumatology, Sagamihara National Hospital, Kanagawa, Japan
| | - Rose Kamenwa
- />Department of Pediatrics and Child Health, Aga Khan University Hospital, Nairobi, Kenya
| | - Bee Wah Lee
- />Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Haiqi Li
- />Department of Primary Child Care, Children’s Hospital, Chongqing Medical University, Chongqing, China
| | - Susan Prescott
- />Department of Immunology, Perth Children’s Hospital, Telethon KIDS Institute, School of Paediatrics and Child Health, University of Western Australia, Crawley, Australia
| | - John J. Riva
- />Department of Clinical Epidemiology & Biostatistics, McMaster University, Hamilton, ON Canada
- />Department of Family Medicine, McMaster University, Hamilton, ON Canada
| | - Lanny Rosenwasser
- />Allergy-Immunology Division, Children’s Mercy Hospital & University of Missouri – Kansas City School of Medicine, Kansas City, MO USA
| | - Hugh Sampson
- />Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | | | - Luigi Terracciano
- />Department of Child and Maternal Medicine, University of Milan Medical School at the Melloni Hospital, Milan, Italy
| | - Andrea Vereda
- />Allergology Department, Hospital Infantil Universitario Niño Jesus, Madrid, Spain
| | - Susan Waserman
- />Department of Medicine, McMaster University, Hamilton, ON Canada
| | - Holger J. Schünemann
- />Department of Clinical Epidemiology & Biostatistics, McMaster University, Hamilton, ON Canada
- />Department of Medicine, McMaster University, Hamilton, ON Canada
| | - Jan L. Brożek
- />Department of Clinical Epidemiology & Biostatistics, McMaster University, Hamilton, ON Canada
- />Department of Medicine, McMaster University, Hamilton, ON Canada
| |
Collapse
|
28
|
Dror T, Dickstein Y, Dubourg G, Paul M. Microbiota manipulation for weight change. Microb Pathog 2016; 106:146-161. [PMID: 26792677 DOI: 10.1016/j.micpath.2016.01.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 01/07/2016] [Accepted: 01/08/2016] [Indexed: 12/21/2022]
Abstract
Manipulation of the intestinal microbiota has been linked to weight changes and obesity. To explore the influence of specific agents that alter the intestinal flora on weight in different patient groups we conducted a meta-analysis of randomized controlled trials (RCTs) reporting on the effects of probiotics, prebiotics, synbiotics, and antibiotics on weight. We searched the Pubmed and Cochrane Library databases for trials on adults, children, and infants evaluating the effects of these substances on weight. Our primary outcome was weight change from baseline. Standardized mean differences (SMDs) with 95% confidence intervals were calculated. We identified and included 13 adult, 17 children, and 23 infant RCTs. Effects were opposite among adults and children, showing weight loss among adults (SMD -0.54 [-0.83, -0.25)) and minor weight gains among children (SMD 0.20 [0.04, 0.36]) and infants (SMD 0.30 [-0.01, 0.62]) taking mainly Lactobacillus probiotic supplements. Heterogeneity was substantial in the adult and infant analyses and could not be explained by intervention or patient characteristics. Azithromycin administration in children with pulmonary disease was associated with weight gain (SMD 0.39 [0.24, 0.54]), without heterogeneity. A high risk of selective reporting and attrition bias was detected across the studies, making it difficult to draw firm conclusions. Overall, our meta-analysis suggests that there may be a role for probiotics in promoting weight loss in adults and weight gain in children, however additional studies are needed. Though we cannot recommend antibiotic administration for weight manipulation, its use provides advantageous weight gain in children with cystic fibrosis and bronchiectasis.
Collapse
Affiliation(s)
- Tal Dror
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Yaakov Dickstein
- Infectious Diseases Institute, Rambam Health Care Campus, Haifa, Israel
| | - Grégory Dubourg
- Pôle des Maladies Infectieuses et Tropicales Clinique et Biologique, Fédération de Bactériologie-Hygiène-Virologie, University, Hospital Centre Timone, Institut Hospitalo-Universitaire (IHU) Méditerranée Infection, Assistance Publique - Hôpitaux de Marseille, Marseille, France; Université Aix-Marseille, Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes (URMITE) UM 63 CNRS 7278 IRD 198 INSERM U1095, Facultés de Médecine et de Pharmacie, Marseille, France
| | - Mical Paul
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel; Infectious Diseases Institute, Rambam Health Care Campus, Haifa, Israel.
| |
Collapse
|
29
|
Munblit D, Boyle RJ, Warner JO. Factors affecting breast milk composition and potential consequences for development of the allergic phenotype. Clin Exp Allergy 2015; 45:583-601. [PMID: 25077553 DOI: 10.1111/cea.12381] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
There is conflicting evidence on the protective role of breastfeeding in relation to allergic sensitization and disease. The factors in breast milk which influence these processes are still unclear and under investigation. We know that colostrum and breast milk contain a variety of molecules which can influence immune responses in the gut-associated lymphoid tissue of a neonate. This review summarizes the evidence that variations in colostrum and breast milk composition can influence allergic outcomes in the infant, and the evidence that maternal and environmental factors can modify milk composition. Taken together, the data presented support the possibility that maternal dietary interventions may be an effective way to promote infant health through modification of breast milk composition.
Collapse
Affiliation(s)
- D Munblit
- Department of Paediatrics, Imperial College London, London, UK; International Inflammation (in-FLAME) Network, of the World Universities Network (WUN)
| | | | | |
Collapse
|
30
|
Abstract
The gastrointestinal (GI) microbiota differs between breast-fed and classic infant formula-fed infants. Breast milk is rich in prebiotic oligosaccharides (OS) and may also contain some probiotics, but scientific societies do not recommend the addition of prebiotic OS or probiotics to standard infant formula. Nevertheless, many infant formula companies often add one or the other or both. Different types of prebiotic OS are used in infant formula, including galacto-oligosaccharide, fructo-oligosaccharide, polydextrose and mixtures of these OS, but none adds human milk OS. There is evidence that the addition of prebiotics to infant formula brings the GI microbiota of formula-fed infants closer to that of breast-fed infants. Prebiotics change gut metabolic activity (by decreasing stool pH and increasing SCFA), have a bifidogenic effect and bring stool consistency and defecation frequency closer to those of breast-fed infants. Although there is only limited evidence that these changes in GI microbiota induce a significant clinical benefit for the immune system, interesting positive trends have been observed in some markers. Additionally, adverse effects are extremely seldom. Prebiotics are added to infant formula because breast milk contains human milk OS. Because most studies suggest a trend of beneficial effects and because these ingredients are very safe, prebiotics bring infant formula one step closer to the golden standard of breast milk.
Collapse
|
31
|
van Stuijvenberg M, Stam J, Grüber C, Mosca F, Arslanoglu S, Chirico G, Braegger CP, Riedler J, Boehm G, Sauer PJJ. Similar Occurrence of Febrile Episodes Reported in Non-Atopic Children at Three to Five Years of Age after Prebiotics Supplemented Infant Formula. PLoS One 2015; 10:e0129927. [PMID: 26076141 PMCID: PMC4468127 DOI: 10.1371/journal.pone.0129927] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 05/04/2015] [Indexed: 12/20/2022] Open
Abstract
This is a follow up study of a multicenter randomised placebo-controlled trial in seven centres in five West European countries. The RCT assessed the effect of infant formula supplemented with a mixture of prebiotics (with neutral short-chain and long-chain oligosaccharides and pectin-derived acidic oligosaccharides) during infancy in term-born children (n=1130). In the follow-up study 672 children (60% of the study population) participated: 232 (56%) from the prebiotics group (PG), 243 (58%) from the control group (CG), and 197 (66%) from the non-randomised breast-fed group (BG). The primary outcome was the occurrence of febrile episodes at three to five years of age prospectively documented by the parents: in the PG 1.17 (interquartile range 0.50-2.08) episodes per year versus 1.20 (0.52-2.57) in the CG; and 1.48 (0.65-2.60) in the BG. This specific prebiotics mixture given during infancy in healthy non-atopic subjects does not decrease febrile episodes and therefore seems not to prevent infection between their third and fifth birthday.
Collapse
Affiliation(s)
- Margriet van Stuijvenberg
- Department of Paediatrics, Beatrix Children’s Hospital, UMC Groningen, Groningen, The Netherlands
- * E-mail:
| | - José Stam
- Department of Paediatrics, Beatrix Children’s Hospital, UMC Groningen, Groningen, The Netherlands
| | - Christoph Grüber
- Department of Pediatric Pulmonology and Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Fabio Mosca
- NICU, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, Milan, Italy
| | - Sertac Arslanoglu
- Centre for Infant Nutrition, Macedonio Melloni Hospital, Milan, Italy
| | | | - Christian P. Braegger
- Division of Pediatric Gastroenterology and Nutrition, University Children’s Hospital, Zürich, Switzerland
| | | | | | - Pieter J. J. Sauer
- Department of Paediatrics, Beatrix Children’s Hospital, UMC Groningen, Groningen, The Netherlands
| | | |
Collapse
|
32
|
Vandenplas Y, Dupont C, Eigenmann P, Host A, Kuitunen M, Ribes-Koninckx C, Shah N, Shamir R, Staiano A, Szajewska H, Von Berg A. A workshop report on the development of the Cow's Milk-related Symptom Score awareness tool for young children. Acta Paediatr 2015; 104:334-9. [PMID: 25557474 DOI: 10.1111/apa.12902] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 11/14/2014] [Accepted: 12/15/2014] [Indexed: 12/17/2022]
Abstract
UNLABELLED Clinicians with expertise in managing children with gastrointestinal problems and/or atopic diseases attended a workshop in Brussels in September 2014 to review the literature and determine whether a clinical score derived from symptoms associated with the ingestion of cow's milk proteins could help primary healthcare providers. The Cow's Milk-related Symptom Score (CoMiSS), which considers general manifestations, dermatological, gastrointestinal and respiratory symptoms, was developed as an awareness tool for cow's milk-related symptoms. It can also be used to evaluate and quantify the evolution of symptoms during therapeutic interventions, but does not diagnose cow's milk protein allergy and does not replace a food challenge. Its usefulness needs to be evaluated by a prospective randomised study. CONCLUSION The CoMiSS provides primary healthcare clinicians with a simple, fast and easy-to-use awareness tool for cow's milk-related symptoms.
Collapse
Affiliation(s)
| | - Christophe Dupont
- Pediatric Gastroenterology; Hepatology and Nutrition Department; Necker Children's Hospital; Paris France
| | - Philippe Eigenmann
- Pediatric Allergy Unit; University Hospitals of Geneva; Geneva Switzerland
| | - Arne Host
- Department of Paediatrics; Hans Christian Andersen Children's Hospital; Odense University Hospital; Odense Denmark
| | - Mikael Kuitunen
- Children's Hospital; University of Helsinki and Helsinki University Central Hospital; Helsinki Finland
| | - Carmen Ribes-Koninckx
- Paediatric Gastroenterology and Hepatology Unit. La Fe University Hospital.; Valencia Spain
| | - Neil Shah
- Great Ormond Street Hospital for Children London (UK) and KU Leuven; Belgium UK
- TARGID; Leuven Belgium
| | - Raanan Shamir
- Institute of Gastroenterology Nutrition and Liver Diseases; Schneider Children's Medical Center of Israel; Sackler Faculty of Medicine Tel-Aviv University; Tel-Aviv Israel
| | - Annamaria Staiano
- Department of Translational Medical Science; Section of Pediatrics; University of Naples Federico II; Naples Italy
| | - Hania Szajewska
- Department of Paediatric; The Medical University of Warsaw; Warsaw Poland
| | - Andrea Von Berg
- Research Institute; Department of Pediatrics; Marien-Hospital; Wesel Germany
| |
Collapse
|
33
|
Abstract
The gastrointestinal microbiota of breast-fed babies differ from classic standard formula fed infants. While mother's milk is rich in prebiotic oligosaccharides and contains small amounts of probiotics, standard infant formula doesn't. Different prebiotic oligosaccharides are added to infant formula: galacto-oligosaccharides, fructo-oligosaccharide, polydextrose, and mixtures of these. There is evidence that addition of prebiotics in infant formula alters the gastrointestinal (GI) microbiota resembling that of breastfed infants. They are added to infant formula because of their presence in breast milk. Infants on these supplemented formula have a lower stool pH, a better stool consistency and frequency and a higher concentration of bifidobacteria in their intestine compared to infants on a non-supplemented standard formula. Since most studies suggest a trend for beneficial clinical effects, and since these ingredients are very safe, prebiotics bring infant formula one step closer to breastmilk, the golden standard. However, despite the fact that adverse events are rare, the evidence on prebiotics of a significant health benefit throughout the alteration of the gut microbiota is limited.
Collapse
Affiliation(s)
- Yvan Vandenplas
- Pediatric Gastroenterology; UZ Brussel; Vrije Universiteit Brussel; Brussels, Belgium,Correspondence to: Yvan Vandenplas;
| | - Elisabeth De Greef
- Pediatric Gastroenterology; UZ Brussel; Vrije Universiteit Brussel; Brussels, Belgium
| | - Gigi Veereman
- Pediatric Gastroenterology; UZ Brussel; Vrije Universiteit Brussel; Brussels, Belgium
| |
Collapse
|
34
|
van de Heijning BJM, Berton A, Bouritius H, Goulet O. GI symptoms in infants are a potential target for fermented infant milk formulae: a review. Nutrients 2014; 6:3942-67. [PMID: 25255831 PMCID: PMC4179197 DOI: 10.3390/nu6093942] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 08/28/2014] [Accepted: 09/09/2014] [Indexed: 12/27/2022] Open
Abstract
Besides pre- and pro-biotic-containing infant formulae, fermented infant formulae are commonly used to relieve or prevent symptoms of gastrointestinal (GI) discomfort in young infants. During the fermentation process in cow's milk-based formulae, the beneficial bacteria modulate the product by forming several beneficial compounds, which contribute to the alleviation of the symptoms observed. This review summarizes the clinical evidence on the impact of fermented infant formulae on common pediatric GI-symptoms. The potential mechanisms involved are discussed: i.e., the lactose and protein (in-) digestibility, effects on gastric emptying and gut transit and modulation of the colonic microbiota. Although initial evidence indicates a beneficial effect of fermented formulae on GI discomfort in newborns, validation and confirmation of the clinical proof obtained so far is warranted, as well as further research to (more fully) understand the mode of action.
Collapse
Affiliation(s)
| | - Amelie Berton
- Nutricia Research, Early Life Nutrition, P.O. Box 80141, 3508 TC Utrecht, The Netherlands.
| | - Hetty Bouritius
- Nutricia Research, Early Life Nutrition, P.O. Box 80141, 3508 TC Utrecht, The Netherlands.
| | - Olivier Goulet
- Department of Pediatric Gastroenterology-Hepatology and Nutrition, Necker Children's Hospital, University of Paris, 75015 Paris, France.
| |
Collapse
|
35
|
Methods to quantify soft tissue-based cranial growth and treatment outcomes in children: a systematic review. PLoS One 2014; 9:e89602. [PMID: 24586904 PMCID: PMC3937373 DOI: 10.1371/journal.pone.0089602] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 01/23/2014] [Indexed: 12/20/2022] Open
Abstract
Context Longitudinal assessment of cranial dimensions of growing children provides healthcare professionals with information about normal and deviating growth as well as treatment outcome. Objective To give an overview of soft tissue–based methods for quantitative longitudinal assessment of cranial dimensions in children until age 6 years and to assess the reliability of these methods in studies with good methodological quality. Data source PubMed, EMBASE, Cochrane Library, Web of Science, Scopus, and CINAHL were searched. A manual search was performed to check for additional relevant studies. Study selection Primary publications on facial growth and treatment outcomes in children younger than age 6 years were included. Data extraction Independent data extraction was performed by two observers. A quality assessment instrument was used to determine methodological quality. Methods used in studies with good methodological quality were assessed for reliability expressed as the magnitude of the measurement error and the correlation coefficient between repeated measurements. Results In total, 165 studies were included, forming three groups of methods: head circumference anthropometry, direct anthropometry, and 2D photography and 3D imaging techniques (surface laser scanning and stereophotogrammetry). In general, the measurement error was below 2 mm, and correlation coefficients were very good. Conclusion Various methods for measuring cranial dimensions have shown to be reliable. Stereophotogrammetry is the most versatile method for quantitative longitudinal assessment of cranial dimensions and shapes in children. However, direct anthropometry continues to be the best method for routine clinical assessments of linear cranial dimensions in growing children until age 6 years.
Collapse
|
36
|
Closa-Monasterolo R, Gispert-Llaurado M, Luque V, Ferre N, Rubio-Torrents C, Zaragoza-Jordana M, Escribano J. Safety and efficacy of inulin and oligofructose supplementation in infant formula: Results from a randomized clinical trial. Clin Nutr 2013; 32:918-27. [DOI: 10.1016/j.clnu.2013.02.009] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Revised: 02/07/2013] [Accepted: 02/13/2013] [Indexed: 12/30/2022]
|
37
|
Mugambi MN, Musekiwa A, Lombard M, Young T, Blaauw R. Association between funding source, methodological quality and research outcomes in randomized controlled trials of synbiotics, probiotics and prebiotics added to infant formula: a systematic review. BMC Med Res Methodol 2013; 13:137. [PMID: 24219082 PMCID: PMC3832685 DOI: 10.1186/1471-2288-13-137] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 11/07/2013] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND There is little or no information available on the impact of funding by the food industry on trial outcomes and methodological quality of synbiotics, probiotics and prebiotics research in infants. The objective of this study was to compare the methodological quality, outcomes of food industry sponsored trials versus non industry sponsored trials, with regards to supplementation of synbiotics, probiotics and prebiotics in infant formula. METHODS A comprehensive search was conducted to identify published and unpublished randomized clinical trials (RCTs). Cochrane methodology was used to assess the risk of bias of included RCTs in the following domains: 1) sequence generation; 2) allocation concealment; 3) blinding; 4) incomplete outcome data; 5) selective outcome reporting; and 6) other bias. Clinical outcomes and authors' conclusions were reported in frequencies and percentages. The association between source of funding, risk of bias, clinical outcomes and conclusions were assessed using Pearson's Chi-square test and the Fisher's exact test. A p-value < 0.05 was statistically significant. RESULTS Sixty seven completed and 3 on-going RCTs were included. Forty (59.7%) were funded by food industry, 11 (16.4%) by non-industry entities and 16 (23.9%) did not specify source of funding. Several risk of bias domains, especially sequence generation, allocation concealment and blinding, were not adequately reported. There was no significant association between the source of funding and sequence generation, allocation concealment, blinding and selective reporting, majority of reported clinical outcomes or authors' conclusions. On the other hand, source of funding was significantly associated with the domains of incomplete outcome data, free of other bias domains as well as reported antibiotic use and conclusions on weight gain. CONCLUSION In RCTs on infants fed infant formula containing probiotics, prebiotics or synbiotics, the source of funding did not influence the majority of outcomes in favour of the sponsors' products. More non-industry funded research is needed to further assess the impact of funding on methodological quality, reported clinical outcomes and authors' conclusions.
Collapse
Affiliation(s)
- Mary N Mugambi
- Division of Human Nutrition, Faculty of Medicine and Health Sciences, Stellenbosch University, P.O Box 19063, Tygerberg 7505, South Africa
| | - Alfred Musekiwa
- Centre for Evidence-Based Health Care, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Martani Lombard
- Division of Human Nutrition, Faculty of Medicine and Health Sciences, Stellenbosch University, P.O Box 19063, Tygerberg 7505, South Africa
| | - Taryn Young
- Centre for Evidence-Based Health Care, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Reneé Blaauw
- Division of Human Nutrition, Faculty of Medicine and Health Sciences, Stellenbosch University, P.O Box 19063, Tygerberg 7505, South Africa
| |
Collapse
|
38
|
Scientific Opinion on nutrient requirements and dietary intakes of infants and young children in the European Union. EFSA J 2013. [DOI: 10.2903/j.efsa.2013.3408] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
39
|
van Goudoever JB, Boehm G. Introduction: bringing science to early life nutrition. Am J Clin Nutr 2013; 98:519S-20S. [PMID: 23824721 DOI: 10.3945/ajcn.112.044164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
40
|
Abstract
BACKGROUND Prebiotics (commonly oligosaccharides) added to infant feeds have the potential to prevent sensitisation of infants to dietary allergens. OBJECTIVES To determine the effect of prebiotic given to infants for the prevention of allergy. SEARCH METHODS We performed an updated search in August 2012 of the Cochrane Central Register of Controlled Trials (CENTRAL) (The Cochrane Library 2012, Issue 8), MEDLINE, EMBASE, conference proceedings, citations, expert informants and clinical trials registries. SELECTION CRITERIA Randomised and quasi-randomised controlled trials that compared the use of a prebiotic to no prebiotic, or a specific prebiotic compared to a different prebiotic in infants for prevention of allergy. DATA COLLECTION AND ANALYSIS Assessment of trial quality, data extraction and synthesis of data were performed using the standard methods of The Cochrane Collaboration. MAIN RESULTS The 2012 update identified 13 studies classified as ongoing or awaiting classification (yet to report allergy outcomes). Forty-three studies were excluded, primarily as no allergy data were reported, although none of these enrolled infants were at high risk of allergy. Four studies enrolling 1428 infants were eligible for inclusion. All studies were at high risk of attrition bias. Allergy outcomes were reported from four months to two years of age.Meta-analysis of two studies (226 infants) found no significant difference in infant asthma although significant heterogeneity was found between studies. Meta-analysis of four studies found a significant reduction in eczema (1218 infants, typical risk ratio 0.68, 95% CI 0.48 to 0.97; typical risk difference -0.04, 95% CI -0.07 to -0.00; number needed to treat to benefit (NNTB) 25, 95% CI 14 to > 100; P = 0.03). No statistically significant heterogeneity was found between studies. One study reported no significant difference in urticaria.No statistically significant subgroup differences were found according to infant risk of allergy or type of infant feed. However, individual studies reported a significant reduction in asthma and eczema from supplementation with a mixture of galacto- and fructo-oligosaccharide (GOS/FOS 9:1 ratio) (8 g/L) in infants at high risk of allergy; and in eczema from supplementation with GOS/FOS (9:1) (6.8 g/L) and acidic oligosacccharide (1.2 g/L) in infants not selected for allergy risk. AUTHORS' CONCLUSIONS Further research is needed before routine use of prebiotics can be recommended for prevention of allergy in formula fed infants. There is some evidence that a prebiotic supplement added to infant feeds may prevent eczema. It is unclear whether the use of prebiotic should be restricted to infants at high risk of allergy or may have an effect in low risk populations; or whether it may have an effect on other allergic diseases including asthma.
Collapse
Affiliation(s)
- David A Osborn
- CentralClinical School,Discipline ofObstetrics,Gynaecology andNeonatology,University of Sydney, Sydney, Australia.
| | | |
Collapse
|