1
|
Barnett D, Thijs C, Mommers M, Endika M, Klostermann C, Schols H, Smidt H, Nauta A, Arts I, Penders J. Why do babies cry? Exploring the role of the gut microbiota in infantile colic, constipation, and cramps in the KOALA birth cohort study. Gut Microbes 2025; 17:2485326. [PMID: 40159147 PMCID: PMC11959906 DOI: 10.1080/19490976.2025.2485326] [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: 10/23/2024] [Revised: 02/17/2025] [Accepted: 03/18/2025] [Indexed: 04/02/2025] Open
Abstract
Gastrointestinal symptoms are common during infancy, including infantile colic. Colic can be loosely defined as prolonged and recurrent crying without obvious cause. The cause indeed remains unclear despite much research. Results on infant nutrition are inconclusive, but prior work has linked maternal mental health to infant crying. Recently, several small studies have described associations between gut microbiota and colic. We used a larger cohort to examine the role of the microbiota in infant gastrointestinal health, while also accounting for other biopsychosocial factors. Using fecal 16S rRNA gene amplicon sequencing data from 1,012 infants in the KOALA birth cohort, we examined associations between the 1-month gut microbiota and parent-reported functional gastrointestinal symptoms throughout infancy, including colic, constipation, and cramps. These analyses were adjusted for biopsychosocial factors that were associated with symptoms in a broader analysis involving 2,665 participants. In 257 infants, we also explored associations between breastmilk human milk oligosaccharides (HMOs) and gastrointestinal symptoms. Higher relative abundance of Staphylococcus at one month was associated with less constipation in the first three months of life. Conversely, Ruminococcus gnavus group abundance was associated with more colicky symptoms, particularly between four and seven months. Breastmilk concentrations of the HMOs lacto-N-hexaose (LNH) and lacto-N-neohexaose (LNnH) were associated with less constipation in the first three months. Our results support the conclusion that gut microbiota are relevant in infantile colic and constipation. However more work is needed to elucidate the underlying mechanisms, and explore their interplay with other relevant biopsychosocial factors such as maternal mental health.
Collapse
Affiliation(s)
- David Barnett
- Maastricht Centre for Systems Biology, Maastricht University, Maastricht, Netherlands
- NUTRIM School for Nutrition and Translational Research in Metabolism, Department of Medical Microbiology, Infectious Diseases and Infection Prevention, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Carel Thijs
- CAPHRI Care and Public Health Research Institute, Department of Epidemiology, Maastricht University, Maastricht, Netherlands
| | - Monique Mommers
- CAPHRI Care and Public Health Research Institute, Department of Epidemiology, Maastricht University, Maastricht, Netherlands
| | - Martha Endika
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, Netherlands
| | - Cynthia Klostermann
- Department of Food Chemistry, Wageningen University & Research, Wageningen, Netherlands
| | - Henk Schols
- Department of Food Chemistry, Wageningen University & Research, Wageningen, Netherlands
| | - Hauke Smidt
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, Netherlands
| | - Arjen Nauta
- FrieslandCampina Ingredients, FrieslandCampina, Amersfoort, Netherlands
| | - Ilja Arts
- Maastricht Centre for Systems Biology, Maastricht University, Maastricht, Netherlands
| | - John Penders
- NUTRIM School for Nutrition and Translational Research in Metabolism, Department of Medical Microbiology, Infectious Diseases and Infection Prevention, Maastricht University Medical Center+, Maastricht, Netherlands
| |
Collapse
|
2
|
Zhen H, Qian H, Liu X, Tan C. Fructooligosaccharides for Relieving Functional Constipation: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Foods 2024; 13:3993. [PMID: 39766936 PMCID: PMC11675838 DOI: 10.3390/foods13243993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/01/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Fructooligosaccharides (FOS) are prebiotics with great potential to improve constipation. This study set out to investigate the usefulness of consuming FOS as a dietary supplement on bowel movement frequency, stool consistency, abundance of Bifidobacteria, gastrointestinal transit time, and gastrointestinal symptoms through a systematic review and meta-analysis of randomized controlled trials (RCTs). We searched the Cochrane Library, PubMed, EMBASE, and Web of Science databases up to 2 March 2024. Randomized controlled trials reporting the use of FOS as a supplement by participants were included. Bias risk was assessed using the Cochrane Risk of Bias assessment tool. Results were synthesized using a random-effects model or fixed-effects model, combining outcomes using odds ratios (OR), weighted mean differences (MD), or standardized mean differences (SMD) with 95% confidence intervals (CI). The meta-analysis encompassed 17 randomized controlled trials, involving a total of 713 study participants. Overall, the intake of FOS significantly increased bowel movement frequency (95% CI: 0.80, 1.50, p < 0.00001) and positively affected stool consistency, softening the stool (SMD: 0.36, 95% CI: 0.12, 0.60, p = 0.76). It also reduced the effort and pain during defecation (SMD: -0.60, 95% CI: -0.85, -0.34, p = 0.12). At the same time, mild bloating was noted as an adverse event associated with FOS ingestion (OR: 10.36, 95% CI: 3.32, 33.23, p = 0.24). No reports of serious adverse events were documented. Overall, FOS may enhance bowel movement frequency, stool consistency, and overall constipation symptoms, suggesting cautious optimism regarding their use as a dietary treatment alternative. Nevertheless, further robust and definitive randomized controlled trials are required to more accurately determine the most effective dosage and duration of use. Additional research and evidence are necessary before the efficacy of FOS as a therapeutic method for treating functional constipation can be firmly established.
Collapse
Affiliation(s)
- Hongmin Zhen
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, China-Canada Joint Lab of Food Nutrition and Health, Beijing Technology and Business University (BTBU), Beijing 100048, China; (H.Z.); (H.Q.)
| | - Hanwei Qian
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, China-Canada Joint Lab of Food Nutrition and Health, Beijing Technology and Business University (BTBU), Beijing 100048, China; (H.Z.); (H.Q.)
| | - Xiaoyan Liu
- China Food Flavor and Nutrition Health Innovation Center, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Chen Tan
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, China-Canada Joint Lab of Food Nutrition and Health, Beijing Technology and Business University (BTBU), Beijing 100048, China; (H.Z.); (H.Q.)
| |
Collapse
|
3
|
Solasaari T, Korpela K, Lommi S, Hyvönen S, Gardemeister S, Merras-Salmio L, Salonen A, de Vos WM, Kolho KL. Bowel function in a prospective cohort of 1052 healthy term infants up to 4 months of age. Eur J Pediatr 2024; 183:3557-3565. [PMID: 38819500 PMCID: PMC11263225 DOI: 10.1007/s00431-024-05625-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/19/2024] [Accepted: 05/22/2024] [Indexed: 06/01/2024]
Abstract
The purpose of this study is to describe the defecation pattern of healthy infants up to 17 weeks of age. We included 1052 healthy term infants from the prospective HELMi cohort (NCT03996304). Parents filled in recurring online questionnaires on feeding, gastrointestinal function, and crying weekly for the first 17 weeks of life. Defecation frequency was highest at the age of 3 weeks (a median of 4 times/day, interquartile range (IQR) 2.9-5). At each time point, the median defecation frequency of breastfed infants was higher than that of infants receiving formula (e.g., at week 17 a median of 2 times/day, IQR 0.9-3.6, and a median of 1.1, IQR 0.6-1.4, respectively). The dominant color of the stool was most often yellow or light brown. Nearly black stools were reported in the first week of life in 3.4%. Nearly half (47.4%) of the infants had green stool color dominating for at least 1 week, with comparable frequency among breastfed (47.7%) and formula-fed (45.2%) infants. Green stools were associated with a higher defecation frequency (linear mixed-effect model p < 0.0001). Occasional blood in stool was reported in 9.3% and recurrent blood in 5.2% of the infants with no difference in stool consistency. Hard stools were rare (≤ 1%). Conclusion: This study enlightens the spectrum of defecation patterns in healthy term infants during the first 17 weeks of life. A better understanding of bowel function helps healthcare professionals distinguish normal from abnormal when addressing defecation, the color of stools, and the type of feeding. What is Known: • Breastfed infants have more frequent and more yellow-colored stools than formula-fed infants. • Stools with green color are often suggested by the parents or even by medical professionals to indicate disease or discomfort in early life. What is New: • Nearly half of the healthy term infants had green stool dominating for at least one week during the first 17 weeks and occasional blood was reported in almost 10% of the infants during this period. • Data on normal variation in bowel function and stool may serve primary health care professionals when educating the families and caretakers of infants.
Collapse
Affiliation(s)
- Terhi Solasaari
- Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Pediatric Clinic, Social Services and Health Care Division, Helsinki, Finland
| | - Katri Korpela
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sohvi Lommi
- Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| | - Sanni Hyvönen
- Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Pediatrics, Tampere University Hospital, Tampere, Finland
| | | | | | - Anne Salonen
- Laboratory of Microbiology, Wageningen University, Wageningen, the Netherlands
| | - Willem M de Vos
- Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Laboratory of Microbiology, Wageningen University, Wageningen, the Netherlands
| | - Kaija-Leena Kolho
- Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Children's Hospital, University of Helsinki and HUS, Helsinki, Finland.
- Faculty of Medicine and Health Technology, University of Tampere, Tampere, Finland.
| |
Collapse
|
4
|
Hou Y, Li J, Ying S. Tryptophan Metabolism and Gut Microbiota: A Novel Regulatory Axis Integrating the Microbiome, Immunity, and Cancer. Metabolites 2023; 13:1166. [PMID: 37999261 PMCID: PMC10673612 DOI: 10.3390/metabo13111166] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 11/16/2023] [Accepted: 11/18/2023] [Indexed: 11/25/2023] Open
Abstract
Tryptophan metabolism and gut microbiota form an integrated regulatory axis that impacts immunity, metabolism, and cancer. This review consolidated current knowledge on the bidirectional interactions between microbial tryptophan processing and the host. We focused on how the gut microbiome controls tryptophan breakdown via the indole, kynurenine, and serotonin pathways. Dysbiosis of the gut microbiota induces disruptions in tryptophan catabolism which contribute to disorders like inflammatory conditions, neuropsychiatric diseases, metabolic syndromes, and cancer. These disruptions affect immune homeostasis, neurotransmission, and gut-brain communication. Elucidating the mechanisms of microbial tryptophan modulation could enable novel therapeutic approaches like psychobiotics and microbiome-targeted dietary interventions. Overall, further research on the microbiota-tryptophan axis has the potential to revolutionize personalized diagnostics and treatments for improving human health.
Collapse
Affiliation(s)
- Yingjian Hou
- Target Discovery Center, China Pharmaceutical University, Nanjing 211198, China;
| | - Jing Li
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha 410000, China
| | - Shuhuan Ying
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
- Shanghai Bocimed Pharmaceutical Research Co., Ltd., Shanghai 201203, China
| |
Collapse
|
5
|
Gao S, Wang J. Maternal and infant microbiome: next-generation indicators and targets for intergenerational health and nutrition care. Protein Cell 2023; 14:807-823. [PMID: 37184065 PMCID: PMC10636639 DOI: 10.1093/procel/pwad029] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/04/2023] [Indexed: 05/16/2023] Open
Abstract
Microbes are commonly sensitive to shifts in the physiological and pathological state of their hosts, including mothers and babies. From this perspective, the microbiome may be a good indicator for diseases during pregnancy and has the potential to be used for perinatal health monitoring. This is embodied in the application of microbiome from multi body sites for auxiliary diagnosis, early prediction, prolonged monitoring, and retrospective diagnosis of pregnancy and infant complications, as well as nutrition management and health products developments of mothers and babies. Here we summarized the progress in these areas and explained that the microbiome of different body sites is sensitive to different diseases and their microbial biomarkers may overlap between each other, thus we need to make a diagnosis prudently for those diseases. Based on the microbiome variances and additional anthropometric and physical data, individualized responses of mothers and neonates to meals and probiotics/prebiotics were predictable, which is of importance for precise nutrition and probiotics/prebiotics managements and developments. Although a great deal of encouraging performance was manifested in previous studies, the efficacy could be further improved by combining multi-aspect data such as multi-omics and time series analysis in the future. This review reconceptualizes maternal and infant health from a microbiome perspective, and the knowledge in it may inspire the development of new options for the prevention and treatment of adverse pregnancy outcomes and bring a leap forward in perinatal health care.
Collapse
Affiliation(s)
- Shengtao Gao
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Jinfeng Wang
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
| |
Collapse
|
6
|
Otten L, Schelker E, Petersen H, Nomayo A, Conzade R, Günther J, Grieger A, Jochum F. Gastrointestinal Tolerance of an Infant Formula Manufactured from Extensively Hydrolysed Protein in Healthy Term Infants. Nutrients 2023; 15:4674. [PMID: 37960327 PMCID: PMC10647512 DOI: 10.3390/nu15214674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 10/24/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
The evaluation of secondary parameters of a prospective, randomised, controlled, multicentre intervention trial aimed to analyse gastrointestinal tolerance of an infant formula manufactured from extensively hydrolysed whey protein (eHF) compared to intact cow's milk protein (control formula, CF) in healthy term infants. Infants ≤ 25 days of age, who were exclusively formula-fed, were randomised to receive eHF or CF for at least three months up to 120 days of age. An exclusively breastfed reference group (BF) was included for descriptive comparison. Infants' gastrointestinal tolerance was evaluated based on stool parameters, the Amsterdam Infant Stool Scale (AISS), the Infant Gastrointestinal Symptom Questionnaire (IGSQ), and sleeping patterns. Of 359 infants included, 297 randomised (eHF: n = 149, CF: n = 148) and 41 BF infants completed the study per protocol. All tolerance parameters were comparable between eHF and CF. Stool was predominantly soft and yellow in colour. Stool was more frequently green in eHF than CF. BF infants had more frequent stools, which were mainly watery or soft and yellow, and comparable IGSQ scores (descriptive). Irrespective of group, all gastrointestinal and sleep parameters showed signs of maturation with increasing age. In conclusion, eHF showed gastrointestinal tolerance as good as CF in healthy infants. Both formulae were well-tolerated.
Collapse
Affiliation(s)
- Lindsey Otten
- Department of Pediatrics, Evangelisches Waldkrankenhaus Spandau, Stadtrandstr. 555, 13589 Berlin, Germany
| | - Elisabeth Schelker
- Department of Pediatrics, Evangelisches Waldkrankenhaus Spandau, Stadtrandstr. 555, 13589 Berlin, Germany
| | - Hanna Petersen
- Department of Pediatrics, Evangelisches Waldkrankenhaus Spandau, Stadtrandstr. 555, 13589 Berlin, Germany
| | - Antonia Nomayo
- Department of Pediatrics, Evangelisches Waldkrankenhaus Spandau, Stadtrandstr. 555, 13589 Berlin, Germany
| | - Romy Conzade
- HiPP GmbH & Co. Vertrieb KG, Georg-Hipp-Str. 7, 85276 Pfaffenhofen an der Ilm, Germany
| | - Julia Günther
- HiPP GmbH & Co. Vertrieb KG, Georg-Hipp-Str. 7, 85276 Pfaffenhofen an der Ilm, Germany
| | - Andrea Grieger
- HiPP GmbH & Co. Vertrieb KG, Georg-Hipp-Str. 7, 85276 Pfaffenhofen an der Ilm, Germany
| | - Frank Jochum
- Department of Pediatrics, Evangelisches Waldkrankenhaus Spandau, Stadtrandstr. 555, 13589 Berlin, Germany
- Department of Pediatrics, Faculty of Medicine, Brandenburg Medical School Theodor Fontane (MHB), Fehrbelliner Str. 38, 16816 Neuruppin, Germany
| | | |
Collapse
|
7
|
Bellaiche M, Tounian P, Oozeer R, Rocher E, Vandenplas Y. Digestive Tolerance and Safety of an Anti-Regurgitation Formula Containing Locust Bean Gum, Prebiotics and Postbiotics: A Real-World Study. Pediatr Gastroenterol Hepatol Nutr 2023; 26:249-265. [PMID: 37736220 PMCID: PMC10509020 DOI: 10.5223/pghn.2023.26.5.249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 06/20/2023] [Accepted: 07/20/2023] [Indexed: 09/23/2023] Open
Abstract
Purpose Infant regurgitation is associated with other functional gastrointestinal disorders and signs and symptoms that have a major impact on the quality of life of infants and their families. This study evaluated the safety, tolerance, and real-world effectiveness of an anti-regurgitation formula containing locust bean gum (LBG), prebiotics, and postbiotics to alleviate digestive symptoms beyond regurgitation. Methods This 3-month study involved infants with regurgitation requiring the prescription of an anti-regurgitation formula according to usual clinical practice. Outcomes included evaluation of the evolution of stool consistency and frequency; occurrence of colic, constipation, and diarrhea; and assessment of regurgitation severity. Infant crying, parental assessment of infant well-being, and parental satisfaction with the stool consistency were also evaluated. Results In total, 190 infants (average age: 1.9±1.1 months) were included. After three months, stool frequency and consistency remained within the normal physiological range, with 82.7% of infants passing one or two stools per day and 90.4% passing loose or formed stools. There was no significant increase in the number of infants with diarrhea, whereas a decrease was observed in the number of infants with constipation after 1 month (p=0.001) and with colic after both 1 and 3 months (p<0.001). Regurgitation severity and crying decreased and parental satisfaction with stool consistency, formula acceptability, infant well-being, and sleep quality increased. Monitoring of adverse events did not reveal any safety concerns. Conclusion Formulas containing LBG, prebiotics, and postbiotics were well tolerated and provided an effective strategy for managing infant regurgitation and gastrointestinal discomfort.
Collapse
Affiliation(s)
- Marc Bellaiche
- Department of Gastroenterology and Pediatric Nutrition, Hospital Robert-Debré, AP-HP, Paris, France
| | - Patrick Tounian
- Department of Pediatric Nutrition and Gastroenterology, Trousseau Hospital, AP-HP, Sorbonne Université, Paris, France
| | | | | | - Yvan Vandenplas
- KidZ Health Castle, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
8
|
Schönknecht YB, Moreno Tovar MV, Jensen SR, Parschat K. Clinical Studies on the Supplementation of Manufactured Human Milk Oligosaccharides: A Systematic Review. Nutrients 2023; 15:3622. [PMID: 37630811 PMCID: PMC10458772 DOI: 10.3390/nu15163622] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/12/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Human milk oligosaccharides (HMOs) are a major component of human milk. They are associated with multiple health benefits and are manufactured on a large scale for their addition to different food products. In this systematic review, we evaluate the health outcomes of published clinical trials involving the supplementation of manufactured HMOs. We screened the PubMed database and Cochrane Library, identifying 26 relevant clinical trials and five publications describing follow-up studies. The clinical trials varied in study populations, including healthy term infants, infants with medical indications, children, and adults. They tested eight different HMO structures individually or as blends in varying doses. All trials included safety and tolerance assessments, and some also assessed growth, stool characteristics, infections, gut microbiome composition, microbial metabolites, and biomarkers. The studies consistently found that HMO supplementation was safe and well tolerated. Infant studies reported a shift in outcomes towards those observed in breastfed infants, including stool characteristics, gut microbiome composition, and intestinal immune markers. Beneficial gut health and immune system effects have also been observed in other populations following HMO supplementation. Further clinical trials are needed to substantiate the effects of HMO supplementation on human health and to understand their structure and dose dependency.
Collapse
|
9
|
Huang M, Shao H, Wang Z, Chen H, Li X. Specific and nonspecific nutritional interventions enhance the development of oral tolerance in food allergy. Crit Rev Food Sci Nutr 2023; 64:10303-10318. [PMID: 37313721 DOI: 10.1080/10408398.2023.2222803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The goal of food allergy (FA) prevention and treatment is to induce oral tolerance (OT). Appropriate nutritional interventions are essential to induce OT to food allergens. This review introduces the mechanism of OT and the importance of early nutritional interventions, and then firstly summarizes specific nutritional factors to induce the development of OT of FA, including proteins, vitamins, fatty acids, saccharides and probiotics. The regulatory mechanism mainly induces the development of tolerance by increasing local or systemic protective regulatory T cells (Tregs) to suppress FA, while the gut microbiota may also be changed to maintain intestinal homeostasis. For allergens-specific OT, the disruption to the structure of proteins and epitopes is critical for the induction of tolerance by hydrolyzed and heated proteins. Vitamins (vitamin A, D), fatty acids, saccharides and probiotics as allergens nonspecific OT also induce the development of OT through immunomodulatory effects. This review contributes to our understanding of OT in FA through nutritional interventions. Nutritional interventions play an important role in the induction of OT, and offer promising approaches to reduce allergy risk and alleviate FA. Moreover, due to the importance and diversity of nutrition, it must be the future trend of induction of OT in FA.
Collapse
Affiliation(s)
- Meijia Huang
- School of Food Science and Technology, Nanchang University, Nanchang, P.R. China
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, P.R. China
| | - Huming Shao
- School of Food Science and Technology, Nanchang University, Nanchang, P.R. China
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, P.R. China
| | - Zhongliang Wang
- School of Food Science and Technology, Nanchang University, Nanchang, P.R. China
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, P.R. China
| | - Hongbing Chen
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, P.R. China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, P.R. China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, P.R. China
| | - Xin Li
- School of Food Science and Technology, Nanchang University, Nanchang, P.R. China
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, P.R. China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, P.R. China
| |
Collapse
|
10
|
Abstract
Abnormalities in gut microbiota have been suggested to be involved in the pathophysiology and progression of Parkinson's disease (PD). Gastrointestinal nonmotor symptoms often precede the onset of motor features in PD, suggesting a role for gut dysbiosis in neuroinflammation and α-synuclein (α-syn) aggregation. In the first part of this chapter, we analyze critical features of healthy gut microbiota and factors (environmental and genetic) that modify its composition. In the second part, we focus on the mechanisms underlying the gut dysbiosis and how it alters anatomically and functionally the mucosal barrier, triggering neuroinflammation and subsequently α-syn aggregation. In the third part, we describe the most common alterations in the gut microbiota of PD patients, dividing the gastrointestinal system in higher and lower tract to examine the association between microbiota abnormalities and clinical features. In the final section, we report on current and future therapeutic approaches to gut dysbiosis aiming to either reduce the risk for PD, modify the disease course, or improve the pharmacokinetic profile of dopaminergic therapies. We also suggest that further studies will be needed to clarify the role of the microbiome in PD subtyping and of pharmacological and nonpharmacological interventions in modifying specific microbiota profiles in individualizing disease-modifying treatments in PD.
Collapse
Affiliation(s)
- Salvatore Bonvegna
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Department of Clinical Neurosciences, Parkinson and Movement Disorders Unit, Milan, Italy
| | - Roberto Cilia
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Department of Clinical Neurosciences, Parkinson and Movement Disorders Unit, Milan, Italy.
| |
Collapse
|
11
|
Murphy S, Carter L, Al Shizawi T, Queally M, Brennan S, O'Neill S. Exploring the relationship between breastfeeding and the incidence of infant illnesses in Ireland: evidence from a nationally representative prospective cohort study. BMC Public Health 2023; 23:140. [PMID: 36670399 PMCID: PMC9854149 DOI: 10.1186/s12889-023-15045-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 01/12/2023] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Ireland has one of the lowest BF rates in the world. This study investigates the association between breastfeeding and infant health in Ireland. METHODS A cross-sectional, secondary analysis of data collected from Growing Up in Ireland (GUI): the National Longitudinal Study of Children was conducted. The average morbidity for 2212. infants exclusively breastfed for at least 90 days (EBF90days) was compared to data for 3987 infants in the non-breastfed (Non-BF) group. Data were weighted using entropy balancing to ensure the comparability of groups. Sensitivity analyses considered alternative definitions of the breastfeeding group. RESULTS Infants who were EBF90days were significantly less likely to be admitted to hospital (CI: - 0.06 to - 0.03), spent less nights in hospital (CI: - 0.37 to - 0.11), and were less likely to develop respiratory diseases including asthma (CI: - 0.03 to - 0.01), chest infections (CI: - 0.12 to - 0.08), snuffles/common colds (CI: - 0.07 to - 0.02), ear infections (CI: - 0.08 to - 0.04), eczema (CI: - 0.08 to - 0.04), skin problems (CI: - 0.04 to - 0.00), wheezing or asthma (CI: - 0.06 to - 0.03), vomiting (CI: - 0.03 to - 0.00), and colic (CI: - 0.04 to - 0.01). Further outcomes such as current health of the infant at time of interview (CI: - 0.04 to - 0.00), feeding problems (CI: - 0.04 to - 0.02) and sleeping problems (CI: - 0.02 to - 0.00) indicated a protective effect of EBF90days versus Non-BF. However, these infants were also more likely to fail to gain weight (CI: 0.01 to 0.02) and were at a slightly higher risk of developing nappy rash (CI: 0.00 to 0.02). CONCLUSION Exclusive breastfeeding for 90+ days is associated with protection against childhood morbidity. Given the protective effect of breastfeeding on adverse health effects in infants, policy makers should prioritise policies that support, promote and protect exclusive breastfeeding.
Collapse
Affiliation(s)
- Sarah Murphy
- School of Medicine, University of Galway, Galway, Ireland
| | - Laura Carter
- J.E. Cairnes School of Business and Economics, University of Galway, Galway, Ireland
| | | | - Michelle Queally
- J.E. Cairnes School of Business and Economics, University of Galway, Galway, Ireland
- Department of Enterprise and Technology, Atlantic Technological University, Galway, Ireland
| | - Sarah Brennan
- Department of General Practice & Donegal Medical Academy, University of Galway, Galway, Ireland.
- General Practice, Carrigart Health Centre, Carrigart, Carrigart, Co. Donegal, Ireland.
| | - Stephen O'Neill
- Department of Health Services Research and Policy, London School of Hygiene and Tropical Medicine, London, UK.
- Centre for Economic and Social Research on Dementia, University of Galway, Galway, Ireland.
| |
Collapse
|
12
|
Lavalle L, Sauvageot N, Cercamondi CI, Egli D, Jankovic I, Vandenplas Y, Juffrie M, Bardosono S, Galindez G, Waheed KAI, Gokhale P, Ibrahim MN, Asghar R, Shirazi H, Perez MLM, Kesavelu D, Edris A, Beleidy AE, Hodhod ME, Elzalabany M, Hussein H, Shaaban SY, Elmashad A, Abdelmoez A, El-Asheer OM, Vandenplas Y, Lavalle L, Sauvageot N, Cercamondi CI, Egli D, Jankovic I, Vandenplas Y. Infant feeding practice and gastrointestinal tolerance: a real-world, multi-country, cross-sectional observational study. BMC Pediatr 2022; 22:714. [PMID: 36514058 DOI: 10.1186/s12887-022-03763-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Signs of feeding intolerance, such as gastrointestinal (GI) symptoms, are frequently observed in otherwise healthy formula-fed infants in the first months of life. The primary objective of this observational study was to examine GI tolerance in formula-fed infants (FFI) vs. breastfed infants (BFI) in a real-world setting with a secondary objective being the comparison of infants fed formula with pre- and/or probiotics (FFI_PP) and those fed formula without any pre- and/or probiotics (FFI_noPP) as well as BFI. METHODS A six-country, cross-sectional study in full-term exclusively/predominantly FFI (n = 2036) and BFI (n = 760) aged 6-16 weeks was conducted using the validated Infant Gastrointestinal Symptom Questionnaire (IGSQ) and a Feeding Practice and Gut Comfort Questionnaire. RESULTS The IGSQ composite score in FFI was non-inferior compared to BFI (mean difference [95%CI]: 0.17 [-0.34, 0.67]; non-inferiority p-value < 0.0001) and scores for BFI and FFI were below the threshold of 23, indicating no GI discomfort. Adjusted mean IGSQ scores ± SE were similar in FFI_PP (22.1 ± 0.2) and BFI (22.3 ± 0.3) while FFI_noPP (23.4 ± 0.3) was significantly higher and above 23 indicating some GI discomfort (mean differences [95%CI] FFI_noPP minus FFI_PP and FFI_noPP minus BFI were 1.28 [0.57, 1.98] and 1.09 [0.38, 1.80], respectively; both p < 0.01). Hard stools and difficulty in passing stool were more common in FFI compared to BFI (p < 0.01) but were less common in FFI_PP compared to FFI_noPP (p < 0.01). FFI_PP showed significantly less crying than FFI_noPP and was similar to BFI. Significantly fewer physician-confirmed colic episodes (Rome IV criteria) were reported in FFI_PP compared with FFI_noPP or BFI. CONCLUSIONS In this real-world observational study, FFI had non-inferior overall GI tolerance compared to BFI. Within FFI, infants receiving formulas with pre- and/or probiotics had a better GI tolerance, improved stooling and less infantile colic compared to those receiving formula without any pre- and/or probiotics and were more similar to BFI. TRIAL REGISTRATION NCT03703583, 12/10/2018 ( https://clinicaltrials.gov/ct2/show/NCT03703583 ).
Collapse
Affiliation(s)
| | - Luca Lavalle
- Biostatistics & Data, Nestlé Research, Lausanne, Switzerland
| | | | - Colin Ivano Cercamondi
- Nestlé Product Technology Center - Nutrition, Société Des Produits Nestlé S.A., 1800, Vevey, Switzerland
| | - Delphine Egli
- Nestlé Product Technology Center - Nutrition, Société Des Produits Nestlé S.A., 1800, Vevey, Switzerland.
| | - Ivana Jankovic
- Nestlé Product Technology Center - Nutrition, Société Des Produits Nestlé S.A., 1800, Vevey, Switzerland
| | - Yvan Vandenplas
- Vrije Universiteit Brussel, UZ Brussels, KidZ Health Castle, Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
van de Heijning BJ, Schoen S, Abrahamse‐Berkeveld M, Hokken‐Koelega AC, van der Beek EM. A concept formula with large, milk phospholipid‐coated lipid droplets enriched with milk fat decreases palmitic acid and calcium levels in stools of healthy, term infants. EUR J LIPID SCI TECH 2022. [DOI: 10.1002/ejlt.202200018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
| | | | | | - Anita C.S. Hokken‐Koelega
- Department of Pediatric Endocrinology Erasmus University Medical Centre – Sophia Children's Hospital
| | | |
Collapse
|
14
|
An Infant Formula with Large, Milk Phospholipid-Coated Lipid Droplets Supports Adequate Growth and Is Well-Tolerated in Healthy, Term Asian Infants: A Randomized, Controlled Double-Blind Clinical Trial. Nutrients 2022; 14:nu14030634. [PMID: 35276993 PMCID: PMC8838783 DOI: 10.3390/nu14030634] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/25/2022] [Accepted: 01/28/2022] [Indexed: 01/06/2023] Open
Abstract
Lipids are essential for healthy infant growth and development. The structural complexity of lipids in human milk is not present in infant milk formula (IF). A concept IF was developed mimicking more closely the structure and composition of human milk fat globules. The current study evaluates whether a concept IF with large, milk phospholipid-coated lipid droplets (mode diameter 3 to 5 μm) is equivalent to standard IF with regard to growth adequacy and safety in healthy, term Asian infants. In this randomized, double-blind, controlled trial, infants were randomized after parents decided to introduce formula. Infants received a standard IF with (Control) or without the specific prebiotic mixture scGOS/lcFOS (9:1 ratio; Control w/o prebiotics), or a Concept IF with large, milk phospholipid-coated lipid droplets and the prebiotic mixture. A group of 67 breastfed infants served as a reference. As a priori defined, only those infants who were fully intervention formula-fed ≤28 days of age were included in the equivalence analysis (Control n = 29; Control w/o prebiotics n = 28; Concept n = 35, per-protocol population). Primary outcome was daily weight gain during the first four months of life, with the difference between the Concept and Control as the key comparison of interest. Additionally, adverse events, growth and tolerance parameters were evaluated. Equivalence of daily weight gain was demonstrated between the Concept and Control group after additional correction for ethnicity and birthweight (difference in estimated means of 0.1 g/d, 90%CI [-2.30, 2.47]; equivalence margin +/- 3 g/d). No clinically relevant group differences were observed in secondary growth outcomes, tolerance outcomes or number, severity or relatedness of adverse events. This study corroborates that an infant formula with large, milk phospholipid-coated lipid droplets supports adequate growth and is well tolerated and safe for use in healthy infants.
Collapse
|
15
|
Infant formula containing bovine milk-derived oligosaccharides supports age-appropriate growth and improves stooling pattern. Pediatr Res 2022; 91:1485-1492. [PMID: 33958719 PMCID: PMC9197766 DOI: 10.1038/s41390-021-01541-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/17/2021] [Accepted: 04/07/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND Adding bovine milk-derived oligosaccharides (MOS) enhances the oligosaccharide profile of infant formula. This study aimed to evaluate the safety and efficacy of a MOS-supplemented infant formula. METHODS In this double-blind randomized controlled trial, healthy infants 21-26 days old were either assigned to bovine milk-based, alpha-lactalbumin, and sn-2 palmitate enriched infant formula (control, n = 115) or the same formula with 7.2 g MOS/L (test, n = 115) until aged 6 months. Co-primary endpoints were weight gain through 4 months and stool consistency (validated scale: 1 = watery to 5 = hard). Secondary endpoints included parent-reported GI tolerance, health-related quality of life (HRQoL), and adverse events (AEs). RESULTS Weight gain was similar (p = 0.695); the difference between test and control (mean; 95% CI: 0.29; -1.15, 1.73 g/day) was above the non-inferiority margin (-3 g/day). Test had softer stools than control (mean difference in stool consistency score: -0.31; 95% CI: -0.42, -0.21; P < 0.0001); fewer parental reports of harder stools (OR = 0.32, 95% CI: 0.20, 0.49; P < 0.0001) and less difficulties in passing stool (OR = 0.25, 95% CI: 0.09, 0.65; P = 0.005). Parent-reported GI tolerance and HRQoL were similar between groups as were the overall low AEs. CONCLUSIONS MOS-supplemented infant formula is safe and well-tolerated while supporting normal infant growth and promotes softer stooling pattern without increasing parent-reported and physician-confirmed adverse health concerns. IMPACT This is the first study investigating the addition of bovine milk-derived oligosaccharides to an infant formula enriched with alpha-lactalbumin and elevated levels of sn-2 palmitate, providing safety and efficacy data for such a formula. Term infant formula supplemented with 7.2 g bovine milk-derived oligosaccharides per liter supported normal infant growth, was well-tolerated and safe. Addition of bovine milk-derived oligosaccharides to term infant formula promoted softer stooling pattern and reduced difficulties in passing stool. The study shows that bovine milk-derived oligosaccharide supplemented infant formula is a safe and effective option for healthy term infants who are formula-fed.
Collapse
|
16
|
Szklany K, Engen PA, Naqib A, Green SJ, Keshavarzian A, Lopez Rincon A, Siebrand CJ, Diks MAP, van de Kaa M, Garssen J, Knippels LMJ, Kraneveld AD. Dietary Supplementation throughout Life with Non-Digestible Oligosaccharides and/or n-3 Poly-Unsaturated Fatty Acids in Healthy Mice Modulates the Gut-Immune System-Brain Axis. Nutrients 2021; 14:173. [PMID: 35011046 PMCID: PMC8746884 DOI: 10.3390/nu14010173] [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: 11/25/2021] [Revised: 12/20/2021] [Accepted: 12/24/2021] [Indexed: 12/11/2022] Open
Abstract
The composition and activity of the intestinal microbial community structures can be beneficially modulated by nutritional components such as non-digestible oligosaccharides and omega-3 poly-unsaturated fatty acids (n-3 PUFAs). These components affect immune function, brain development and behaviour. We investigated the additive effect of a dietary combination of scGOS:lcFOS and n-3 PUFAs on caecal content microbial community structures and development of the immune system, brain and behaviour from day of birth to early adulthood in healthy mice. Male BALB/cByJ mice received a control or enriched diet with a combination of scGOS:lcFOS (9:1) and 6% tuna oil (n-3 PUFAs) or individually scGOS:lcFOS (9:1) or 6% tuna oil (n-3 PUFAs). Behaviour, caecal content microbiota composition, short-chain fatty acid levels, brain monoamine levels, enterochromaffin cells and immune parameters in the mesenteric lymph nodes (MLN) and spleen were assessed. Caecal content microbial community structures displayed differences between the control and dietary groups, and between the dietary groups. Compared to control diet, the scGOS:lcFOS and combination diets increased caecal saccharolytic fermentation activity. The diets enhanced the number of enterochromaffin cells. The combination diet had no effects on the immune cells. Although the dietary effect on behaviour was limited, serotonin and serotonin metabolite levels in the amygdala were increased in the combination diet group. The combination and individual interventions affected caecal content microbial profiles, but had limited effects on behaviour and the immune system. No apparent additive effect was observed when scGOS:lcFOS and n-3 PUFAs were combined. The results suggest that scGOS:lcFOS and n-3 PUFAs together create a balance-the best of both in a healthy host.
Collapse
Affiliation(s)
- Kirsten Szklany
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands; (K.S.); (A.L.R.); (C.J.S.); (M.A.P.D.); (M.v.d.K.); (J.G.); (L.M.J.K.)
| | - Phillip A. Engen
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush Medical College, Rush University Medical Center, Chicago, IL 60602, USA; (P.A.E.); (A.N.); (A.K.)
| | - Ankur Naqib
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush Medical College, Rush University Medical Center, Chicago, IL 60602, USA; (P.A.E.); (A.N.); (A.K.)
| | - Stefan J. Green
- Genomics and Microbiome Core Facility, Rush University Medical Center, Chicago, IL 60602, USA;
| | - Ali Keshavarzian
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush Medical College, Rush University Medical Center, Chicago, IL 60602, USA; (P.A.E.); (A.N.); (A.K.)
- Department of Medicine & Physiology, Rush University Medical Center, Chicago, IL 60602, USA
| | - Alejandro Lopez Rincon
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands; (K.S.); (A.L.R.); (C.J.S.); (M.A.P.D.); (M.v.d.K.); (J.G.); (L.M.J.K.)
- Department of Data Science, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, 3584 EA Utrecht, The Netherlands
| | - Cynthia J. Siebrand
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands; (K.S.); (A.L.R.); (C.J.S.); (M.A.P.D.); (M.v.d.K.); (J.G.); (L.M.J.K.)
| | - Mara A. P. Diks
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands; (K.S.); (A.L.R.); (C.J.S.); (M.A.P.D.); (M.v.d.K.); (J.G.); (L.M.J.K.)
| | - Melanie van de Kaa
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands; (K.S.); (A.L.R.); (C.J.S.); (M.A.P.D.); (M.v.d.K.); (J.G.); (L.M.J.K.)
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands; (K.S.); (A.L.R.); (C.J.S.); (M.A.P.D.); (M.v.d.K.); (J.G.); (L.M.J.K.)
- Global Centre of Excellence Immunology, Nutricia Danone Research, 3584 CT Utrecht, The Netherlands
| | - Leon M. J. Knippels
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands; (K.S.); (A.L.R.); (C.J.S.); (M.A.P.D.); (M.v.d.K.); (J.G.); (L.M.J.K.)
- Global Centre of Excellence Immunology, Nutricia Danone Research, 3584 CT Utrecht, The Netherlands
| | - Aletta D. Kraneveld
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands; (K.S.); (A.L.R.); (C.J.S.); (M.A.P.D.); (M.v.d.K.); (J.G.); (L.M.J.K.)
| |
Collapse
|
17
|
Bellaiche M, Ludwig T, Arciszewska M, Bongers A, Gomes C, Świat A, Dakhlia F, Piollet A, Oozeer R, Vandenplas Y. Safety and Tolerance of a Novel Anti-Regurgitation Formula: A Double-Blind, Randomized, Controlled Trial. J Pediatr Gastroenterol Nutr 2021; 73:579-585. [PMID: 34417399 PMCID: PMC8528134 DOI: 10.1097/mpg.0000000000003289] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 05/29/2021] [Indexed: 01/02/2023]
Abstract
OBJECTIVES A novel anti-regurgitation (AR) formula has been designed to support gut health and improve gastrointestinal (GI) symptoms beyond regurgitation. This study assessed the tolerance and safety of this new AR formula. METHODS This was a 4-week double-blind, randomized, controlled trial with a 4-week extension in formula-fed infants with regurgitation. The new AR (Test) formula contained 0.4 g/100 mL locust bean gum (LBG) as thickener, partly fermented formula with postbiotics, and short-chain galacto-oligosaccharides (scGOS) and long-chain fructo-oligosaccharides (lcFOS) (0.4 g/100 mL, ratio 9:1). The Control AR formula contained LBG (0.4 g/100 mL) with postbiotics and has a history of safe use. The primary outcome was the Infant Gastrointestinal Symptom Questionnaire (IGSQ) sum score including stooling, spitting-up/vomiting, crying, fussiness and flatulence. RESULTS All 182 infants screened were enrolled in the study. The primary analysis showed the equivalence of the IGSQ sum scores at Week 4 between groups. IGSQ sum scores improved significantly within 1 week (Mixed Model Repeated Measurement [MMRM], P < 0.001). Post-hoc analyses showed a bigger improvement of the IGSQ score in the Test (n = 38) versus Control (n = 44) group (MMRM, P = 0.008) in infants with more severe gastrointestinal (GI) symptoms (IGSQ score ≥35). Stool characteristics were comparable between groups. Growth related z scores were in line with the WHO child growth standards and both groups showed improvement of regurgitation. Adverse events did not show any safety concerns. CONCLUSIONS The novel AR formula combining LBG, scGOS/lcFOS and postbiotics is well-tolerated, safe and supports adequate growth during the intervention. Post-hoc analyses suggest that the formula results in more improvement of GI symptom burden in infants with more severe symptoms.
Collapse
Affiliation(s)
| | | | | | - Anke Bongers
- Danone Nutricia Research, Utrecht, The Netherlands
| | | | - Agnieszka Świat
- Centrum Medyczne Promed
- Centrum Medyczne Plejady, Krakow, Poland
| | - Faouzi Dakhlia
- Centre Condorcet, Hôpital Privé d’Antony, Antony, France
| | | | - Raish Oozeer
- Danone Nutricia Research, Utrecht, The Netherlands
| | - Yvan Vandenplas
- KidZ Health Castle, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
18
|
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: 0.8] [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
|
19
|
Vandenplas Y, Gerlier L, Caekelbergh K, Possner M. An Observational Real-Life Study with a New Infant Formula in Infants with Functional Gastro-Intestinal Disorders. Nutrients 2021; 13:nu13103336. [PMID: 34684337 PMCID: PMC8539302 DOI: 10.3390/nu13103336] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/15/2021] [Accepted: 09/18/2021] [Indexed: 12/28/2022] Open
Abstract
Functional gastro-intestinal disorders (FGIDs) impair the quality of life of many infants and their families. A formula with partial whey hydrolysate, starch, high magnesium content, prebiotic fructo-oligosaccharide and galacto-oligosaccharide and the probiotic Lactobacillus reuteri DSM 17938 was given during two weeks to 196 infants with at least two FGIDs. The efficacy was evaluated with the Cow Milk-associated Symptom Score (CoMiSS®) and quality of life with the QUALIN score. The formula was shown to decrease FGIDs within three days (decrease of CoMiSS −1.29 (3.15) (mean (SD), p < 0.0001) followed by an improvement of quality of life after seven days (increase QUALIN +1.4 (7.8); p: 0.008). Constipation decreased from 18.8% to 6.5% within three days. In combination with reassurance and guidance, the nutritional intervention was shown to be effective in infants with FGIDS in real-life circumstances.
Collapse
Affiliation(s)
- Yvan Vandenplas
- KidZ Health Castle, Vrije Universiteit Brussel (VUB), UZ Brussel, 1090 Brussels, Belgium
- Correspondence: ; Tel.: +32-475748794
| | | | | | | | - Mike Possner
- Nestlé Nutrition Institute, 60528 Frankfurt am Main, Germany;
| |
Collapse
|
20
|
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: 1.3] [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
|
21
|
Long-Term Safety and Efficacy of Prebiotic Enriched Infant Formula-A Randomized Controlled Trial. Nutrients 2021; 13:nu13041276. [PMID: 33924514 PMCID: PMC8070502 DOI: 10.3390/nu13041276] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/07/2021] [Accepted: 04/09/2021] [Indexed: 01/15/2023] Open
Abstract
The present study aims to evaluate the effects of an infant formula supplemented with a mixture of prebiotic short and long chain inulin-type oligosaccharides on health outcomes, safety and tolerance, as well as on fecal microbiota composition during the first year of life. In a prospective, multicenter, randomized, double-blind study, n = 160 healthy term infants under 4 months of age were randomized to receive either an infant formula enriched with 0.8 g/dL of Orafti®Synergy1 or an unsupplemented control formula until the age of 12 months. Growth, fever (>38 °C) and infections were regularly followed up by a pediatrician. Digestive symptoms, stool consistency as well as crying and sleeping patterns were recorded during one week each study month. Fecal microbiota and immunological biomarkers were determined from a subgroup of infants after 2, 6 and 12 months of life. The intention to treat (ITT) population consisted of n = 149 infants. Both formulae were well tolerated. Mean duration of infections was significantly lower in the prebiotic fed infants (p < 0.05). The prebiotic group showed higher Bifidobacterium counts at month 6 (p = 0.006), and higher proportions of Bifidobacterium in relation to total bacteria at month 2 and 6 (p = 0.042 and p = 0.013, respectively). Stools of infants receiving the prebiotic formula were softer (p < 0.05). Orafti®Synergy1 tended to beneficially impact total daily amount of crying (p = 0.0594). Supplementation with inulin-type prebiotic oligosaccharides during the first year of life beneficially modulates the infant gut microbiota towards higher Bifidobacterium levels at the first 6 months of life, and is associated with reduced duration of infections.
Collapse
|
22
|
Ludwig T, Oukid I, Wong J, Ting S, Huysentruyt K, Roy P, Foussat AC, Vandenplas Y. Machine Learning Supports Automated Digital Image Scoring of Stool Consistency in Diapers. J Pediatr Gastroenterol Nutr 2021; 72:255-261. [PMID: 33275399 PMCID: PMC7815249 DOI: 10.1097/mpg.0000000000003007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/09/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND/AIMS Accurate stool consistency classification of non-toilet-trained children remains challenging. This study evaluated the feasibility of automated classification of stool consistencies from diaper photos using machine learning (ML). METHODS In total, 2687 usable smartphone photos of diapers with stool from 96 children younger than 24 months were obtained after independent ethical study approval. Stool consistency was assessed from each photo according to the original 7 types of the Brussels Infant and Toddler Stool Scale independently by study participants and 2 researchers. A health care professional assigned a final score in case of scoring disagreement between the researchers. A proof-of-concept ML model was built upon this collected photo database, using transfer learning to re-train the classification layer of a pretrained deep convolutional neural network model. The model was built on random training (n = 2478) and test (n = 209) subsets. RESULTS Agreements between study participants and both researchers were 58.0% and 48.5%, respectively, and between researchers 77.5% (assessable n = 2366). The model classified 60.3% of the test photos in exact agreement with the final score. With respect to the 4-class grouping of the 7 Brussels Infant and Toddler Stool Scale types, the agreement between model-based and researcher classification was 77.0%. CONCLUSION The automated and objective scoring of stool consistency from diaper photos by the ML model shows robust agreement with human raters and overcomes limitations of other methods relying on caregiver reporting. Integrated with a smartphone application, this new framework for photo database construction and ML classification has numerous potential applications in clinical studies and home assessment.
Collapse
Affiliation(s)
- Thomas Ludwig
- Danone Nutricia Research, Precision Nutrition D-lab, Biopolis, Singapore
| | | | - Jill Wong
- Danone Nutricia Research, Precision Nutrition D-lab, Biopolis, Singapore
| | - Steven Ting
- Danone Nutricia Research, Precision Nutrition D-lab, Biopolis, Singapore
| | - Koen Huysentruyt
- KidZ Health Castle, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Puspita Roy
- Danone Nutricia Research, Precision Nutrition D-lab, Biopolis, Singapore
| | - Agathe C. Foussat
- Danone Nutricia Research, Precision Nutrition D-lab, Biopolis, Singapore
| | - Yvan Vandenplas
- KidZ Health Castle, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
23
|
Rani L, Mondal AC. Unravelling the role of gut microbiota in Parkinson's disease progression: Pathogenic and therapeutic implications. Neurosci Res 2021; 168:100-112. [PMID: 33417973 DOI: 10.1016/j.neures.2021.01.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 01/02/2021] [Accepted: 01/03/2021] [Indexed: 02/07/2023]
Abstract
In recent years, researchers have shown interest in bi-directional interaction between the brain and gut, called "gut-brain axis". Emerging pieces of evidence indicate that disturbances in this axis is found to be associated with the Parkinson's disease (PD). Several clinical investigations revealed the crucial role of gut microbiota in the pathogenesis of PD. It has been suggested that aggregation of misfolded protein α-syn, the neuropathological hallmark of PD, might begin in gut and propagates to the CNS via vagus nerve and olfactory bulb. Emerging evidences also suggest that initiation and progression of PD may be due to inflammation originating from gut. It has been shown that microbial gut dysbiosis causes the production of various pathogenic microbial metabolites which elevates pro-inflammatory environment in the gut that promotes neuroinflammation in the CNS. These observations raise the intriguing question - how gut microbial dysbiosis could contribute to PD progression. In this context, various microbiota-targeted therapies are under consideration that can re-establish the intestinal homeostasis which may have greater promise in the prevention and treatment of PD. This review focuses on the role of the gut microbiota in the initiation, progression of PD and current therapeutic intervention to deplete the severity of the disease.
Collapse
Affiliation(s)
- Linchi Rani
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, Delhi, India
| | - Amal Chandra Mondal
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, Delhi, India.
| |
Collapse
|
24
|
Vandenplas Y, de Halleux V, Arciszewska M, Lach P, Pokhylko V, Klymenko V, Schoen S, Abrahamse-Berkeveld M, Mulder KA, Porcel Rubio R. A Partly Fermented Infant Formula with Postbiotics Including 3'-GL, Specific Oligosaccharides, 2'-FL, and Milk Fat Supports Adequate Growth, Is Safe and Well-Tolerated in Healthy Term Infants: A Double-Blind, Randomised, Controlled, Multi-Country Trial. Nutrients 2020; 12:nu12113560. [PMID: 33233658 PMCID: PMC7699816 DOI: 10.3390/nu12113560] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/12/2020] [Accepted: 11/16/2020] [Indexed: 12/21/2022] Open
Abstract
This study investigated growth, safety, and tolerance in healthy infants consuming a partly fermented infant formula (IF) with postbiotics, 2′-linked fucosyllactose (2′-FL), a specific prebiotic mixture of short-chain galacto-oligosaccharides (scGOS) and long-chain fructo-oligosaccharides (lcFOS), and milk fat. This double-blind, controlled trial randomised 215 fully IF-fed infants ≤ 14 days of age to either: Test Group (IF) containing 26% fermented formula with postbiotics derived from Lactofidus fermentation process (including 3′-Galactosyllactose; 3′-GL), 0.8 g/100 mL scGOS/lcFOS (9:1), 0.1 g/100 mL 2′-FL, and milk fat), or Control group (IF with 0.8 g/100 mL scGOS/lcFOS (9:1)) until 17 weeks of age. Fully breastfed infants were included as a reference. Anthropometric measures, gastrointestinal symptoms, and safety were assessed monthly. Equivalence in weight gain (primary outcome) between the Test and Control groups was confirmed (difference in means −0.08 g/day; 90% CI (−1.47;1.31)) with estimated mean weight gain (SE) of 31.00 (0.59) g/day and 31.08 (0.60) g/day, respectively, (PP population, n = 196). Equivalence in length and head circumference gain between the randomised groups was also confirmed. No statistically significant differences were observed in adverse events or gastrointestinal tolerance between randomised IF groups. A partly fermented IF with postbiotics, specific oligosaccharides, 2′-FL, and milk fat supports adequate infant growth and is safe and well-tolerated in healthy term infants.
Collapse
Affiliation(s)
- Yvan Vandenplas
- KidZ Health Castle, UZ Brussel, Vrije Universiteit Brussel, 1090 Jette, Belgium
- Correspondence: ; Tel.: +32-02-477-5794
| | - Virginie de Halleux
- Centre Hospitalier Universitaire of Liège (CHU), Centre Hospitalier Régional of Liège (CHR), 4000 Liège, Belgium;
| | | | - Piotr Lach
- Centrum Medyczne Promed, 31-411 Kraków, Poland;
| | - Valeriy Pokhylko
- Ukrainian Medical Stomatological Academy, Poltava Regional Children Clinical Hospital, 36011 Poltava, Ukraine;
| | - Viktoriia Klymenko
- Kharkiv National Medical University, City Children’s Clinical Hospital No. 19, 61051 Kharkiv, Ukraine;
| | - Stefanie Schoen
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.S.); (M.A.-B.); (K.A.M.)
| | | | - Kelly A Mulder
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.S.); (M.A.-B.); (K.A.M.)
| | | |
Collapse
|
25
|
An Infant Formula with Partially Hydrolyzed Whey Protein Supports Adequate Growth and Is Safe and Well-Tolerated in Healthy, Term Infants: A Randomized, Double-Blind, Equivalence Trial. Nutrients 2020; 12:nu12072072. [PMID: 32668666 PMCID: PMC7400250 DOI: 10.3390/nu12072072] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/06/2020] [Accepted: 07/09/2020] [Indexed: 12/02/2022] Open
Abstract
The current study evaluates the safety and tolerance of a partially hydrolyzed whey protein-based infant formula (PHF) versus an in intact cow’s milk protein formula (IPF). Breastfed infants were included as a reference group. In a multi-country, multicenter, randomized, double-blinded, controlled clinical trial, infants whose mothers intended to fully formula feed were randomized to PHF (n = 134) or IPF (n = 134) from ≤14 days to 17 weeks of age. The equivalence analysis of weight gain per day within margins of +/−3 g/d (primary outcome), the recorded adverse events, growth and gastro-intestinal tolerance parameters were considered for the safety evaluation. Equivalence of weight gain per day from enrolment until 17 weeks of age was demonstrated in the PHF group compared to the IPF group (difference in means −1.2 g/d; 90% CI (−2.42; 0.02)), with estimated means (SE) of 30.2 (0.5) g/d and 31.4 (0.5) g/d, respectively. No significant differences in growth outcomes, the number, severity or type of (serious) adverse events and tolerance outcomes, were observed between the two formula groups. A partially hydrolyzed whey protein-based infant formula supports adequate infant growth, with a daily weight gain equivalent to a standard intact protein-based formula; it is also safe for use and well-tolerated in healthy term infants.
Collapse
|
26
|
Steutel NF, Zeevenhooven J, Scarpato E, Vandenplas Y, Tabbers MM, Staiano A, Benninga MA. Prevalence of Functional Gastrointestinal Disorders in European Infants and Toddlers. J Pediatr 2020; 221:107-114. [PMID: 32446468 DOI: 10.1016/j.jpeds.2020.02.076] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/29/2020] [Accepted: 02/27/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To assess the prevalence of functional gastrointestinal disorders (FGIDs) in young children in a cross-sectional, multicenter study in Belgium, Italy, and The Netherlands. STUDY DESIGN Children were enrolled if they were age 0-48 months, attending a general pediatrician (Belgium, Italy) or a well-baby clinic (The Netherlands) for routine follow-up. Separate questionnaires were developed for infants age 0-12 months and for toddlers age 13-48 months. Questionnaires evaluated the clinical history, symptoms, sociodemographic information on the family, and exposure to stressful life events. FGIDs were defined according to Rome IV criteria. RESULTS In total 2751 children were included: 1698 infants age 0-12 months and 1053 children age 13-48 months. The prevalence of any FGID in infants age 0-12 months and 13-48 months was 24.7% and 11.3%, respectively. The most common disorders were infant regurgitation (13.8%) in infants and functional constipation (9.6%) in toddlers. Multivariable regression analyses demonstrated that younger age (P = .030) and formula feeding (P = .045) were associated with the prevalence of any FGID among infants. Country (Italy) (P = .033) and parents subjected to domestic violence (P = .035) were associated with the prevalence of any FGID in toddlers age 13-48 months. CONCLUSIONS FGIDs are common in a community sample of Western European infants and toddlers. Regurgitation is most prevalent in infants and functional constipation is most common in toddlers. Younger age, formula feeding, and domestic violence to parents are associated with the prevalence of FGIDs.
Collapse
Affiliation(s)
- Nina F Steutel
- Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Pediatric Gastroenterology, Hepatology and Nutrition, Amsterdam, The Netherlands; Amsterdam UMC, University of Amsterdam, Clinical Epidemiology, Bioinformatics and Biostatistics, Amsterdam, The Netherlands
| | - Judith Zeevenhooven
- Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Pediatric Gastroenterology, Hepatology and Nutrition, Amsterdam, The Netherlands
| | - Elena Scarpato
- Department of Translational Medical Sciences, Section of Pediatrics, University of Naples Federico II, Naples, Italy
| | - Yvan Vandenplas
- KidZ Health Castle, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Merit M Tabbers
- Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Pediatric Gastroenterology, Hepatology and Nutrition, Amsterdam, The Netherlands
| | - Annamaria Staiano
- Department of Translational Medical Sciences, Section of Pediatrics, University of Naples Federico II, Naples, Italy
| | - Marc A Benninga
- Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Pediatric Gastroenterology, Hepatology and Nutrition, Amsterdam, The Netherlands
| |
Collapse
|
27
|
Bondue P, Milani C, Arnould E, Ventura M, Daube G, LaPointe G, Delcenserie V. Bifidobacterium mongoliense genome seems particularly adapted to milk oligosaccharide digestion leading to production of antivirulent metabolites. BMC Microbiol 2020; 20:111. [PMID: 32380943 PMCID: PMC7206731 DOI: 10.1186/s12866-020-01804-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 04/26/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Human milk oligosaccharides (HMO) could promote the growth of bifidobacteria, improving young children's health. In addition, fermentation of carbohydrates by bifidobacteria can result in the production of metabolites presenting an antivirulent activity against intestinal pathogens. Bovine milk oligosaccharides (BMO), structurally similar to HMO, are found at high concentration in cow whey. This is particularly observed for 3'-sialyllactose (3'SL). This study focused on enzymes and transport systems involved in HMO/BMO metabolism contained in B. crudilactis and B. mongoliense genomes, two species from bovine milk origin. The ability of B. mongoliense to grow in media supplemented with whey or 3'SL was assessed. Next, the effects of cell-free spent media (CFSM) were tested against the virulence expression of Escherichia coli O157:H7 and Salmonella enterica serovar Typhimurium. RESULTS Due to the presence of genes encoding β-galactosidases, β-hexosaminidases, α-sialidases and α-fucosidases, B. mongoliense presents a genome more sophisticated and more adapted to the digestion of BMO/HMO than B. crudilactis (which contains only β-galactosidases). In addition, HMO/BMO digestion involves genes encoding oligosaccharide transport systems found in B. mongoliense but not in B. crudilactis. B. mongoliense seemed able to grow on media supplemented with whey or 3'SL as main source of carbon (8.3 ± 1.0 and 6.7 ± 0.3 log cfu/mL, respectively). CFSM obtained from whey resulted in a significant under-expression of ler, fliC, luxS, stx1 and qseA genes (- 2.2, - 5.3, - 2.4, - 2.5 and - 4.8, respectively; P < 0.05) of E. coli O157:H7. CFSM from 3'SL resulted in a significant up-regulation of luxS (2.0; P < 0.05) gene and a down-regulation of fliC (- 5.0; P < 0.05) gene. CFSM obtained from whey resulted in significant up-regulations of sopD and hil genes (2.9 and 3.5, respectively; P < 0.05) of S. Typhimurium, while CFSM obtained from 3'SL fermentation down-regulated hil and sopD genes (- 2.7 and - 4.2, respectively; P < 0.05). CONCLUSION From enzymes and transporters highlighted in the genome of B. mongoliense and its potential ability to metabolise 3'SL and whey, B. mongoliense seems well able to digest HMO/BMO. The exact nature of the metabolites contained in CFSM has to be identified still. These results suggest that BMO associated with B. mongoliense could be an interesting synbiotic formulation to maintain or restore intestinal health of young children.
Collapse
Affiliation(s)
- Pauline Bondue
- Department of Food Science, Fundamental and Applied Research for Animal and Health, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Christian Milani
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Emilie Arnould
- Department of Food Science, Fundamental and Applied Research for Animal and Health, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Georges Daube
- Department of Food Science, Fundamental and Applied Research for Animal and Health, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Gisèle LaPointe
- Canadian Research Institute for Food Safety, University of Guelph, Guelph, Canada
| | - Véronique Delcenserie
- Department of Food Science, Fundamental and Applied Research for Animal and Health, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium.
| |
Collapse
|
28
|
Gazerani P. Probiotics for Parkinson's Disease. Int J Mol Sci 2019; 20:E4121. [PMID: 31450864 PMCID: PMC6747430 DOI: 10.3390/ijms20174121] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 08/14/2019] [Accepted: 08/21/2019] [Indexed: 02/08/2023] Open
Abstract
Parkinson's disease (PD) is a complex neurological disorder classically characterized by impairments in motor system function associated with loss of dopaminergic neurons in the substantia nigra. After almost 200 years since the first description of PD by James Parkinson, unraveling the complexity of PD continues to evolve. It is now recognized that an interplay between genetic and environmental factors influences a diverse range of cellular processes, reflecting on other clinical features including non-motor symptoms. This has consequently highlighted the extensive value of early clinical diagnosis to reduce difficulties of later stage management of PD. Advancement in understanding of PD has made remarkable progress in introducing new tools and strategies such as stem cell therapy and deep brain stimulation. A link between alterations in gut microbiota and PD has also opened a new line. Evidence exists of a bidirectional pathway between the gastrointestinal tract and the central nervous system. Probiotics, prebiotics and synbiotics are being examined that might influence gut-brain axis by altering gut microbiota composition, enteric nervous system, and CNS. This review provides status on use of probiotics for PD. Limitations and future directions will also be addressed to promote further research considering use of probiotics for PD.
Collapse
Affiliation(s)
- Parisa Gazerani
- Biomedicine: Department of Health Science and Technology, Faculty of Medicine, Aalborg University,Frederik Bajers Vej 3B, 9220 Aalborg East, Denmark.
| |
Collapse
|
29
|
Gastrointestinal Tolerance, Growth and Safety of a Partly Fermented Formula with Specific Prebiotics in Healthy Infants: A Double-Blind, Randomized, Controlled Trial. Nutrients 2019; 11:nu11071530. [PMID: 31284424 PMCID: PMC6683277 DOI: 10.3390/nu11071530] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/01/2019] [Accepted: 07/03/2019] [Indexed: 12/14/2022] Open
Abstract
This study evaluated the effect of a partly fermented infant formula (using the bacterial strains Bifidobacterium breve C50 and Streptococcus thermophilus 065) with a specific prebiotic mixture (short-chain galacto-oligosaccharides (scGOS) and long-chain fructo-oligosaccharides (lcFOS; 9:1)) on the incidence of gastrointestinal symptoms, stool characteristics, sleeping and crying behaviour, growth adequacy and safety. Two-hundred infants ≤28 days of age were assigned either to experimental infant formula containing 30% fermented formula and 0.8 g/100 mL scGOS/lcFOS or to non-fermented control infant formula without scGOS/lcFOS. A group of breastfed infants served as a reference. No relevant differences in parent-reported gastrointestinal symptoms were observed. Stool consistency was softer in the experimental versus control group with values closer to the breastfed reference group. Daily weight gain was equivalent for both formula groups (0.5 SD margins) with growth outcomes close to breastfed infants. No clinically relevant differences in adverse events were observed, apart from a lower investigator-reported prevalence of infantile colic in the experimental versus control group (1.1% vs. 8.7%; p < 0.02). Both study formulae are well-tolerated, support an adequate infant growth and are safe for use in healthy term infants. Compared to the control formula, the partly fermented formula with prebiotics induces stool consistencies closer to breastfed infants.
Collapse
|
30
|
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: 1.0] [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
|
31
|
Breij LM, Abrahamse-Berkeveld M, Vandenplas Y, Jespers SNJ, de Mol AC, Khoo PC, Kalenga M, Peeters S, van Beek RHT, Norbruis OF, Schoen S, Acton D, Hokken-Koelega ACS. An infant formula with large, milk phospholipid-coated lipid droplets containing a mixture of dairy and vegetable lipids supports adequate growth and is well tolerated in healthy, term infants. Am J Clin Nutr 2019; 109:586-596. [PMID: 30793165 PMCID: PMC6408203 DOI: 10.1093/ajcn/nqy322] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 10/16/2018] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Lipid droplets in human milk have a mode diameter of ∼4 μm and are surrounded by a native phospholipid-rich membrane. Current infant milk formulas (IMFs) contain small lipid droplets (mode diameter ∼0.5 μm) primarily coated by proteins. A concept IMF was developed mimicking more closely the structure and composition of human milk lipid droplets. OBJECTIVES This randomized, controlled, double-blind equivalence trial evaluates the safety and tolerance of a concept IMF with large, milk phospholipid-coated lipid droplets (mode diameter 3-5 μm) containing vegetable and dairy lipids in healthy, term infants. METHODS Fully formula-fed infants were enrolled up to 35 d of age and randomly assigned to 1 of 2 formulas until 17 wk of age: 1) Control IMF with small lipid droplets containing vegetable oils (n = 108); or 2) Concept IMF with large, milk phospholipid-coated lipid droplets comprised of 48% dairy lipids (n = 115). A group of 88 breastfed infants served as reference. Primary outcome was daily weight gain during intervention. Additionally, number and type of adverse events, growth, and tolerance parameters were monitored. RESULTS Equivalence of daily weight gain was demonstrated (Concept compared with Control IMF: -1.37 g/d; 90% CI: -2.71, -0.02; equivalence margin ± 3 g/d). No relevant group differences were observed in growth, tolerance and number, severity, or relatedness of adverse events. We did observe a higher prevalence of watery stools in the Concept than in the Control IMF group between 5 and 12 wk of age (P < 0.001), closer to the stool characteristics observed in the breastfed group. CONCLUSIONS An infant formula with large, milk phospholipid-coated lipid droplets containing dairy lipids is safe, well tolerated, and supports an adequate growth in healthy infants. This trial was registered in the Dutch Trial Register (www.trialregister.nl) as NTR3683.
Collapse
Affiliation(s)
- Laura M Breij
- Erasmus University Medical Centre/Sophia Children's Hospital, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | - Anita C S Hokken-Koelega
- Erasmus University Medical Centre/Sophia Children's Hospital, Rotterdam, The Netherlands,Address correspondence to AH-K (e-mail: )
| | | |
Collapse
|
32
|
Randomized, Double-Blind, Placebo-Controlled Parallel Clinical Trial Assessing the Effect of Fructooligosaccharides in Infants with Constipation. Nutrients 2018; 10:nu10111602. [PMID: 30388751 PMCID: PMC6266108 DOI: 10.3390/nu10111602] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 10/25/2018] [Accepted: 10/26/2018] [Indexed: 01/29/2023] Open
Abstract
Constipation often begins in the first year of life. The aim of this study was to assess the effect of fructooligosaccharides (FOS) in the treatment of infants with constipation. This randomized, double-blind, placebo-controlled clinical trial included infants with constipation who were randomly assigned to one of two parallel groups: FOS or placebo. Either the FOS supplement or the placebo was added to the infant formula. Thirty-six infants completed the 4-week intervention. Therapeutic success occurred in 83.3% of the FOS group infants and in 55.6% of the control group infants (p = 0.073; one-tailed test). Compared with the control group, the FOS group exhibited a higher frequency of softer stools (p = 0.035) and fewer episodes of straining and/or difficulty passing stools (p = 0.041). At the end of the intervention, the mouth-to-anus transit time was shorter (22.4 and 24.5 h, p = 0.035), and the Bifidobacterium sp. count was higher (p = 0.006) in the FOS group. In conclusion, the use of FOS in infants with constipation was associated with significant improvement in symptoms, but the results showed no statistical significance regarding the success of the therapy compared with the control group. FOS was associated with reduced bowel transit time and higher counts of the genus Bifidobacterium in the stool.
Collapse
|
33
|
Infant Feeding Practices and the Effect in Reducing Functional Constipation 6 Years Later: A Randomized Field Trial. J Pediatr Gastroenterol Nutr 2018; 67:660-665. [PMID: 29927865 DOI: 10.1097/mpg.0000000000002075] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
OBJECTIVE The aim of this study is to assess the impact of health worker training on infant feeding practices on the prevalence of functional constipation (FC) among children at 6 years of age. METHODS Cluster randomized field trial conducted in Porto Alegre, Brazil. Health centers were randomly allocated into intervention (n = 9) or control (n = 11) groups. In intervention sites, health workers joined training sessions on the "Ten Steps for Healthy Feeding for Children from Birth to Two Years of age". Pregnant women in the last trimester of both groups were identified, invited to participate and enrolled in the study as the potential mothers to receive the dietary counseling provided by the health workers. At 6 years of age, the prevalence of FC was evaluated based on Rome III, defined by 2 or more of the following: infrequent defecation, fecal incontinence, history of retentive posturing, or/and history of painful defecation. RESULTS Among 387 mother-child pairs (206 intervention, 181 control) evaluated at 6 years of age, the prevalence of FC was lower in the intervention group compared with the control group (15.0% vs 23.9%, respectively). The probability of being constipated was 38% lower in the intervention group (PR = 0.62; 95% CI 0.44-0.87; P < 0.01). CONCLUSION The health workers training to promote the "Ten Steps" was an effective way to reduce the prevalence of constipation among children at 6 years of age.
Collapse
|
34
|
Nijman RM, Liu Y, Bunyatratchata A, Smilowitz JT, Stahl B, Barile D. Characterization and Quantification of Oligosaccharides in Human Milk and Infant Formula. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:6851-6859. [PMID: 29799744 DOI: 10.1021/acs.jafc.8b01515] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Oligosaccharides are known to affect the health of infants. The analysis of these complex molecules in (human) milk samples requires state-of-the-art techniques. This study analyzed the composition and concentration of oligosaccharides in early (day 3) and mature (day 42) human milk as well as in five different infant formula brands. The oligosaccharide content decreased in human milk from 9.15 ± 0.25 g/L at day 3 to 6.38 ± 0.29 g/L at day 42 of lactation. All formulas resulted to be fortified with galacto-oligosaccharides, with one also fortified with polydextrose and another with long-chain fructo-oligosaccharides. About 130 unique oligosaccharide structures were identified in the human milk samples, whereas infant formula contained less diversity of structures. The comparisons indicated that composition and abundance of oligosaccharides unique to human milk are not yet reproduced in infant formulas. The analytical workflow developed is suitable for the determination of prebiotic oligosaccharides in foods that contain diverse carbohydrate structures.
Collapse
Affiliation(s)
- Rose M Nijman
- Danone Nutricia Research , 3584 CT Utrecht , Netherlands
| | - Yan Liu
- Department of Food Science and Technology , University of California, Davis , Davis , California 95616 , United States
| | - Apichaya Bunyatratchata
- Department of Food Science and Technology , University of California, Davis , Davis , California 95616 , United States
| | - Jennifer T Smilowitz
- Department of Food Science and Technology , University of California, Davis , Davis , California 95616 , United States
| | - Bernd Stahl
- Danone Nutricia Research , 3584 CT Utrecht , Netherlands
| | - Daniela Barile
- Department of Food Science and Technology , University of California, Davis , Davis , California 95616 , United States
| |
Collapse
|
35
|
Common gastrointestinal distress among infants: Role of optimal nutritional interventions. CLINICAL EPIDEMIOLOGY AND GLOBAL HEALTH 2018. [DOI: 10.1016/j.cegh.2017.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
36
|
Abstract
The market of functional foods has experienced a huge growth in the last decades due to the increased consumers’ awareness in a healthy lifestyle. Dried fruits constitute good snacks, in alternative to salty or sweet ones, and food ingredients due to their taste and nutritional/health benefits. Bioactive molecules are interesting sources to develop functional foods, as they play a major role in improving the health status and minimizing disease risks. The bioactive compounds most widely discussed in literature are presented in this review, for example, polyphenols, phytosterols, and prebiotics. Different technologies to dry bioproducts for producing functional foods or ingredients are presented. New drying techniques for the preservation of bioactive compounds are proposed, focusing more specifically on dielectric drying. A discussion on the techniques that can be used to optimize drying processes is performed. An overview on dehydrated plant based foods with probiotics is provided. The microorganisms used, impregnation procedures, drying methods, and evaluated parameters are presented and discussed. The principal bioactive compounds responsible for nutritional and health benefits of plant derived dried food products—fruits and vegetables, fruits and vegetables by-products, grains, nuts, and algae—are presented. Phytochemical losses occurring during pretreatments and/or drying processes are also discussed.
Collapse
|
37
|
Perez-Pardo P, Kliest T, Dodiya HB, Broersen LM, Garssen J, Keshavarzian A, Kraneveld AD. The gut-brain axis in Parkinson's disease: Possibilities for food-based therapies. Eur J Pharmacol 2017; 817:86-95. [DOI: 10.1016/j.ejphar.2017.05.042] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 03/31/2017] [Accepted: 05/22/2017] [Indexed: 12/18/2022]
|
38
|
Vandenplas Y, Ludwig T, Bouritius H, Alliet P, Forde D, Peeters S, Huet F, Hourihane J. Randomised controlled trial demonstrates that fermented infant formula with short-chain galacto-oligosaccharides and long-chain fructo-oligosaccharides reduces the incidence of infantile colic. Acta Paediatr 2017; 106:1150-1158. [PMID: 28328065 PMCID: PMC5485044 DOI: 10.1111/apa.13844] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 11/28/2016] [Accepted: 03/17/2017] [Indexed: 12/20/2022]
Abstract
AIM We examined the effects on gastrointestinal (GI) tolerance of a novel infant formula that combined specific fermented formula (FERM) with short-chain galacto-oligosaccharides and long-chain fructo-oligosaccharides (scGOS/lcFOS), with a 9:1 ratio and concentration of 0.8 g/100 mL. METHODS This prospective, double-blind, randomised, controlled trial comprised 432 healthy, term infants aged 0-28 days whose parents decided to not start, or discontinued, breastfeeding. Infant formula with scGOS/lcFOS+50%FERM, scGOS/lcFOS+15%FERM, 50%FERM and scGOS/lcFOS were tested. Parents completed standardised seven-day diaries on GI symptoms, crying, sleeping and stool characteristics each month until the infants were 17 weeks. RESULTS All the formulas were well tolerated. At four weeks, the overall incidence of infantile colic was significantly lower (8%) with scGOS/lcFOS+50%FERM than scGOS/lcFOS (20%, p = 0.034) or 50%FERM (20%, p = 0.036). Longitudinal modelling showed that scGOS/lcFOS+50%FERM-fed infants also displayed a persistently lower daily crying duration and showed a consistent stool-softening effect than infants who received formula without scGOS/lcFOS. CONCLUSION The combination of fermented formula with scGOS/lcFOS was well tolerated and showed a lower overall crying time, a lower incidence of infantile colic and a stool-softening effect in healthy term infants. These findings suggest for the first time that a specific infant formula has a preventive effect on infantile colic in formula-fed infants.
Collapse
Affiliation(s)
- Yvan Vandenplas
- Department of Pediatrics; UZ, Vrije Universiteit Brussel; Brussels Belgium
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Perez-Pardo P, de Jong EM, Broersen LM, van Wijk N, Attali A, Garssen J, Kraneveld AD. Promising Effects of Neurorestorative Diets on Motor, Cognitive, and Gastrointestinal Dysfunction after Symptom Development in a Mouse Model of Parkinson's Disease. Front Aging Neurosci 2017; 9:57. [PMID: 28373840 PMCID: PMC5357625 DOI: 10.3389/fnagi.2017.00057] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 02/27/2017] [Indexed: 01/13/2023] Open
Abstract
Parkinson's disease (PD) is characterized by the progressive degeneration of dopaminergic nigrostriatal neurons, with reductions in the function and amount of dopaminergic synapses. Therefore, synapse loss and membrane-related pathology provide relevant targets for interventions in PD. We previously showed the beneficial preventive effects of a dietary intervention containing uridine and DHA, two precursors for membrane synthesis, in the intrastriatal rotenone model for PD. Here, we examined the therapeutic potential of the same dietary intervention on motor, cognitive, and gastrointestinal symptoms. In addition, we tested the effects of an extended nutritional formula based on the same precursors plus other nutrients that increase membrane phospholipid synthesis as well as prebiotic fibers. C57BL/6J mice received a unilateral rotenone injection in the striatum. Dietary interventions started 28 days after surgery, when motor-symptoms had developed. Readout parameters included behavioral tasks measuring motor function and spatial memory as well as intestinal function and histological examination of brain and gut to assess PD-like pathology. Our results show that rotenone-induced motor and non-motor problems were partially alleviated by the therapeutic dietary interventions providing uridine and DHA. The extended nutritional intervention containing both precursors and other nutrients that increase phospholipid synthesis as well as prebiotic fibers was more effective in normalizing rotenone-induced motor and non-motor abnormalities. The latter diet also restored striatal DAT levels, indicating its neurorestorative properties. This is the first study demonstrating beneficial effects of specific dietary interventions, given after full development of symptoms, on a broad spectrum of motor and non-motor symptoms in a mouse model for PD.
Collapse
Affiliation(s)
- Paula Perez-Pardo
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University Utrecht, Netherlands
| | - Esther M de Jong
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University Utrecht, Netherlands
| | - Laus M Broersen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht UniversityUtrecht, Netherlands; Nutricia ResearchUtrecht, Netherlands
| | | | | | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht UniversityUtrecht, Netherlands; Nutricia ResearchUtrecht, Netherlands
| | - Aletta D Kraneveld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University Utrecht, Netherlands
| |
Collapse
|
40
|
Ribeiro Vieira C, Laurides Ribeiro de Oliveira Lomeu F, de Castro Moreira ME, Stampini Duarte Martino H, Ribeiro Silva R. Clinical application of a cocoa and unripe banana flour beverage for overweight women with abdominal obesity: Prospective, double-blinded and randomized clinical trial. J Food Biochem 2017. [DOI: 10.1111/jfbc.12372] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
41
|
Partly Fermented Infant Formulae With Specific Oligosaccharides Support Adequate Infant Growth and Are Well-Tolerated. J Pediatr Gastroenterol Nutr 2016; 63:e43-53. [PMID: 27472478 PMCID: PMC5051523 DOI: 10.1097/mpg.0000000000001360] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Fermented formulae (FERM) and a specific mixture of 90% short-chain galacto-oligosaccharides and 10% long-chain fructo-oligosaccharides (scGOS/lcFOS; 9:1) have a potential beneficial effect on gastrointestinal function and microbiota development in infants. The present study assessed the safety and tolerance of the combination of partly fermented infant milk formulae and scGOS/lcFOS compared with either 1 feature, in healthy term infants. METHODS Four hundred thirty-two infants were enrolled before 28 days of age and followed up to 17 weeks of age and assigned to 1 of the 4 groups: (i) formula with scGOS/lcFOS, (ii) scGOS/lcFOS + 15% FERM, (iii) scGOS/lcFOS + 50% FERM, or (iv) 50% fermented formula (50% FERM). Primary outcome was daily weight gain during intervention (equivalence criterion: difference in daily weight gain ≤3 g/day). Infants' anthropometrics, formula intake, number, and type of (serious) AEs were monitored monthly. Stool samples were collected at baseline and after 17 weeks for analysis of physiological and microbiological parameters. RESULTS Equivalence of weight gain per day was demonstrated in both the intention-to-treat and per-protocol population, with a mean weight gain (SD) of 29.7 (6.1), 28.2 (4.8), 28.5 (5.0), and 28.7 (5.9) g/day for the groups i to iv respectively. No differences were observed in other growth parameters, formula intake, and the number or severity of AEs. In all scGOS/lcFOS-containing formulae, a beneficial effect of scGOS/lcFOS was observed, indicated by the lower pH, lower Clostridium difficile levels, and higher secretory immunoglobulin A levels. CONCLUSIONS The partly fermented infant milk formulae containing the specific mixture scGOS/lcFOS were well-tolerated and resulted in normal growth in healthy infants.
Collapse
|
42
|
Bondue P, Crèvecoeur S, Brose F, Daube G, Seghaye MC, Griffiths MW, LaPointe G, Delcenserie V. Cell-Free Spent Media Obtained from Bifidobacterium bifidum and Bifidobacterium crudilactis Grown in Media Supplemented with 3'-Sialyllactose Modulate Virulence Gene Expression in Escherichia coli O157:H7 and Salmonella Typhimurium. Front Microbiol 2016; 7:1460. [PMID: 27713728 PMCID: PMC5031695 DOI: 10.3389/fmicb.2016.01460] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 09/01/2016] [Indexed: 11/19/2022] Open
Abstract
Complex oligosaccharides from human milk (HMO) possess an antimicrobial activity and can promote the growth of bifidobacteria such as Bifidobacterium bifidum and Bifidobacterium longum subsp. infantis. In addition, fermentation of carbohydrates by bifidobacteria can result in the production of metabolites presenting an antivirulence effect on several pathogenic bacteria. Whey is rich in complex bovine milk oligosaccharides (BMO) structurally similar to HMO and B. crudilactis, a species of bovine origin, is able to metabolize some of those complex carbohydrates. This study focused on the ability of B. bifidum and B. crudilactis to grow in a culture medium supplemented in 3′-sialyllactose (3′SL) as the main source of carbon, a major BMO encountered in cow milk. Next, the effects of cell-free spent media (CFSM) were tested against virulence expression of Escherichia coli O157:H7 and Salmonella enterica serovar Typhimurium. Both strains were able to grow in presence of 3′SL, but B. crudilactis showed the best growth (7.92 ± 0.3 log cfu/ml) compared to B. bifidum (6.84 ± 0.9 log cfu/ml). Then, CFSM were tested for their effects on virulence gene expression by ler and hilA promoter activity of luminescent mutants of E. coli and S. Typhimurium, respectively, and on wild type strains of E. coli O157:H7 and S. Typhimurium using RT-qPCR. All CFSM resulted in significant under expression of the ler and hilA genes for the luminescent mutants and ler (ratios of −15.4 and −8.1 respectively) and qseA (ratios of −2.1 and −3.1) for the wild type strain of E. coli O157:H7. The 3′SL, a major BMO, combined with some bifidobacteria strains of bovine or human origin could therefore be an interesting synbiotic to maintain or restore the intestinal health of young children. These effects observed in vitro will be further investigated regarding the overall phenotype of pathogenic agents and the exact nature of the active molecules.
Collapse
Affiliation(s)
- Pauline Bondue
- Department of Food Science, Fundamental and Applied Research for Animal and Health, Faculty of Veterinary Medicine, University of Liège Liège, Belgium
| | - Sébastien Crèvecoeur
- Department of Food Science, Fundamental and Applied Research for Animal and Health, Faculty of Veterinary Medicine, University of Liège Liège, Belgium
| | - François Brose
- Department of Food Science, Fundamental and Applied Research for Animal and Health, Faculty of Veterinary Medicine, University of Liège Liège, Belgium
| | - Georges Daube
- Department of Food Science, Fundamental and Applied Research for Animal and Health, Faculty of Veterinary Medicine, University of Liège Liège, Belgium
| | | | - Mansel W Griffiths
- Canadian Research Institute for Food Safety, University of Guelph Guelph, Canada
| | - Gisèle LaPointe
- Canadian Research Institute for Food Safety, University of Guelph Guelph, Canada
| | - Véronique Delcenserie
- Department of Food Science, Fundamental and Applied Research for Animal and Health, Faculty of Veterinary Medicine, University of Liège Liège, Belgium
| |
Collapse
|
43
|
Signs and symptoms associated with digestive tract development. J Pediatr (Rio J) 2016; 92:S46-56. [PMID: 27020622 DOI: 10.1016/j.jped.2016.02.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 02/24/2016] [Accepted: 02/20/2016] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE To analyze the development and prevalence of gastrointestinal signs and symptoms associated with the development of the digestive tract, and to assess the measures aimed to reduce their negative impacts. SOURCE OF DATA Considering the scope and comprehensiveness of the subject, a systematic review of the literature was not carried out. The Medline database was used to identify references that would allow the analysis of the study topics. SYNTHESIS OF RESULTS Infants frequently show several gastrointestinal signs and symptoms. These clinical manifestations can be part of gastrointestinal functional disorders such as infantile colic, infant regurgitation, and functional constipation. Allergy to cow's milk protein and gastroesophageal reflux disease are also causes of these clinical manifestations and represent an important and difficult differential diagnosis. The diseases that course with gastrointestinal signs and symptoms can have an impact on family dynamics and maternal emotional status, and may be associated with future problems in the child's life. Comprehensive pediatric care is essential for diagnosis and treatment. Maternal breastfeeding should always be maintained. Some special formulas can contribute to the control of clinical manifestations depending on the established diagnosis. CONCLUSION During the normal development of the digestive tract, several gastrointestinal signs and symptoms may occur, usually resulting from functional gastrointestinal disorders, gastroesophageal reflux disease, and allergy to cow's milk protein. Breastfeeding should always be maintained.
Collapse
|
44
|
de Morais MB. Signs and symptoms associated with digestive tract development. JORNAL DE PEDIATRIA (VERSÃO EM PORTUGUÊS) 2016. [DOI: 10.1016/j.jpedp.2016.03.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
45
|
Abrahamse E, Huybers S, Alles MS, Renes IB, Knol J, Bouritius H, Ludwig T. Fermented Infant Formula Increases Ileal Protein Digestibility and Reduces Ileal Proteolytic Activity Compared with Standard and Hydrolyzed Infant Formulas in Piglets. J Nutr 2015; 145:1423-8. [PMID: 26019249 DOI: 10.3945/jn.114.208314] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 04/23/2015] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND An infant formula that contained milk fermented by the bacteria Bifidobacterium breve and Streptococcus thermophilus (Lactofidus) was reported to alleviate functional digestive symptoms in infants. It was hypothesized that improved protein digestibility of the fermented infant formula could contribute to this effect. OBJECTIVE The aim of this study was to evaluate the protein digestibility of a specific fermented (FF), a standard (SF), and an extensively hydrolyzed protein (HF) formula. METHODS Four-week-old piglets (n = 7) were fitted with a T-cannula at the terminal ileum and received each formula in a Latin square design. FF, SF, and HF contained 11.7%, 9.3%, and 11.9% (w/w) crude protein; 1.5%, 5.4%, and 5.6% (w/w) fiber; and had a casein/whey ratio of 60:40, 50:50, and 0:100 per kilogram of powder, respectively. Ileal digesta were collected and analyzed for amino acids and proteolytic activity. RESULTS FF had a significantly higher apparent ileal crude protein digestibility (92.1% ± 1.0%) than SF and HF (84.4% ± 1.0% and 83.9% ± 0.9%, respectively). FF also had a significantly higher dry matter digestibility than SF and HF. The ileal crude protein flow of FF was significantly lower than that of SF and HF. The ileal flow of FF total proteolytic activity was significantly lower than that of SF but not significantly different from that of HF (412 ± 163 kU/8 h vs. 1530 ± 163 and 703 ± 156 kU/8 h, respectively). CONCLUSIONS The FF in piglets had a significantly higher apparent ileal crude protein digestibility than the SF and HF and displayed lower ileal proteolytic activity than the SF. Both effects may contribute to the alleviation of functional gastrointestinal symptoms reported in infants fed fermented infant milk formula.
Collapse
Affiliation(s)
- Evan Abrahamse
- Department of Developmental Physiology and Nutrition, Nutricia Research, Utrecht, The Netherlands
| | - Sylvie Huybers
- Department of Developmental Physiology and Nutrition, Nutricia Research, Utrecht, The Netherlands
| | - Martine S Alles
- Department of Developmental Physiology and Nutrition, Nutricia Research, Utrecht, The Netherlands
| | - Ingrid B Renes
- Department of Developmental Physiology and Nutrition, Nutricia Research, Utrecht, The Netherlands
| | - Jan Knol
- Department of Developmental Physiology and Nutrition, Nutricia Research, Utrecht, The Netherlands
| | - Hetty Bouritius
- Department of Developmental Physiology and Nutrition, Nutricia Research, Utrecht, The Netherlands
| | - Thomas Ludwig
- Department of Developmental Physiology and Nutrition, Nutricia Research, Utrecht, The Netherlands
| |
Collapse
|
46
|
Bondue P, Delcenserie V. Genome of Bifidobacteria and Carbohydrate Metabolism. Korean J Food Sci Anim Resour 2015; 35:1-9. [PMID: 26761794 PMCID: PMC4682508 DOI: 10.5851/kosfa.2015.35.1.1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 02/08/2015] [Indexed: 11/25/2022] Open
Abstract
In recent years, the knowledge about bifidobacteria has considerably evolved thanks to recent progress in molecular biology. The analysis of the whole genome sequences of 48 taxa of bifidobacteria offers new perspectives for their classification, especially to set up limit between two species. Indeed, several species are presenting a high homology and should be reclassified. On the other hand, some subspecies are presenting a low homology and should therefore be reclassified into different species. In addition, a better knowledge of the genome of bifidobacteria allows a better understanding of the mechanisms involved in complex carbohydrate metabolism. The genome of some species of bifidobacteria from human but also from animal origin demonstrates high presence in genes involved in the metabolism of complex oligosaccharides. Those species should be further tested to confirm their potential to metabolize complex oligosaccharides in vitro and in vivo.
Collapse
Affiliation(s)
- Pauline Bondue
- Fundamental and Applied Research for Animal & Health (FARAH), Food Science Department, Faculty of Veterinary Medicine, University of Liège, Sart-Tilman, B43b Liege, B-4000 Belgium
| | - Véronique Delcenserie
- Fundamental and Applied Research for Animal & Health (FARAH), Food Science Department, Faculty of Veterinary Medicine, University of Liège, Sart-Tilman, B43b Liege, B-4000 Belgium
| |
Collapse
|