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] [Download PDF] [Figures] [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
|
Li H, He B, Ma N, Liu C, Cai K, Zhang X, Ma X. Quorum sensing of Bifidobacteria: Research and progress. Microbiol Res 2025; 294:128102. [PMID: 39965277 DOI: 10.1016/j.micres.2025.128102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 01/17/2025] [Accepted: 02/12/2025] [Indexed: 02/20/2025]
Abstract
Quorum sensing (QS) is a common method of communication among bacteria. While previous studies have discovered the mechanisms of QS in a variety of pathogenic bacteria, relatively little research has focused on probiotics, such as Bifidobacteria. Recent studies have detected QS signalling molecules in Bifidobacteria, but it remains unclear whether the probiotic properties of Bifidobacteria are mediated by QS. This review aims to provide an overview of the QS system in Bifidobacteria and its role in promoting the secretion of metabolites such as extracellular vesicles and biofilms. The review further examines the inhibition of virulence gene expression by Bifidobacteria QS through the luxS/AI-2 system, as well as its role in promoting host-microbial interactions. Understanding the QS mechanisms of Bifidobacteria can reveal beneficial interactions with hosts, which may facilitate the control of bacterial infections, including therapeutic strategies for intestinal diseases. This knowledge can also help improve gut health, thereby addressing the opportunities and challenges of enhancing the body's nutritional status.
Collapse
Affiliation(s)
- Huahui Li
- College of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Bin He
- College of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Ning Ma
- College of Animal Science and Technology, China Agricultural University, Haidian, Beijing 100193, China
| | - Chunchen Liu
- College of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Kun Cai
- College of Animal Science and Technology, China Agricultural University, Haidian, Beijing 100193, China
| | - Xiujun Zhang
- College of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China.
| | - Xi Ma
- College of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China; College of Animal Science and Technology, China Agricultural University, Haidian, Beijing 100193, China.
| |
Collapse
|
3
|
Brasiel PGDA, Potente Dutra Luquetti SC. Effects of Probiotics Supplementation on Short-Chain Fatty Acids: A Systematic Review of Randomized Controlled Trials. Nutr Rev 2025:nuaf047. [PMID: 40265671 DOI: 10.1093/nutrit/nuaf047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025] Open
Abstract
CONTEXT With the increased use of fermented products and probiotic foods, interest in knowing their benefits and safety of their intake is increasing. OBJECTIVE The current systematic review investigated the effects of probiotics supplementation on short-chain fatty acid (SCFA) levels. METHODS A systematic review was conducted according to the PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines. Medline/PubMed, Scopus, Web of Science, and Embase databases were searched from inception to February 2024, including only randomized controlled trials. RESULTS A total of 30 studies were included, involving 1499 participants. The vast majority of trials investigated Bifidobacterium and Lactobacillus strains. These were categorized into healthy adults (n = 6), gastrointestinal/inflammatory diseases (n = 7), metabolic diseases (n = 6), elderly individuals (n = 3), children (n = 4), and infant formula (n = 4). Most studies evaluated isolated strain supplementation (n = 12), followed by fermented beverages (n = 11) and probiotic mixes (n = 3). Globally, 16 studies (53.3%) revealed an increase in at least 1 SCFA in participants supplemented with probiotics. In comparison, 5 studies (16.7%) reported a reduction and 9 studies showed no statistically significant impact in their findings (30%). The subgroup evaluation showed heterogeneity in the results, with low to moderate evidence grading levels. CONCLUSION Probiotics supplementation during childhood was the intervention period more effective in increasing fecal SCFAs and immunoglobulin A (IgA) levels (GRADE: moderate/high-certainty evidence). SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CRD42024513221.
Collapse
|
4
|
Brown JM, Baran JV, Lamos L, Beacker J, Florio J, Oliveros LV, Fabbrini AL, Farrar AA, Wilsey MJ. Infant Colic Symptoms and Amino Acid Formula: Insights from a Prospective Cohort Study. Nutrients 2025; 17:1302. [PMID: 40284167 PMCID: PMC12029966 DOI: 10.3390/nu17081302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/17/2025] [Accepted: 03/31/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Infant functional disorders, including colic, irritability, and sleep disturbances, often overlap with symptoms of cow's milk protein allergy (CMPA). While extensively hydrolyzed formulas are commonly used to address these issues, the short-term efficacy of amino acid formulas (AAF) for managing colic remains understudied. This secondary analysis of a previously published prospective cohort, the largest of its kind in the United States, evaluated the short-term impact of AAF in improving colic and associated symptoms in infants under six months of age with suspected CMPA. Methods: This real-world prospective cohort study analyzed data from 138 infants with suspected CMPA initiated on AAF at Visit 1. After excluding 34 infants due to incomplete data or treatment changes, 104 infants were included in the final analysis. Symptom severity and resolution were assessed through outcomes across two visits, with care provided by general pediatricians (82%) and pediatric gastroenterologists (18%). Results: At baseline, the most common symptoms were colic (n = 83), gassiness (n = 72), fussiness (n = 66), and sleep disturbances (n = 58). By the next follow-up visit (Visit 2), parents reported symptom improvements in colic (94%), gassiness (81%), fussiness (83%), and sleep disturbances (86%). Complete symptom resolution was observed in 73% of infants with colic, 68% with gassiness, 65% with fussiness, and 81% with sleep difficulties. By Visit 2, no infants had severe symptoms, with only mild residual symptoms reported. Conclusions: AAF was associated with significant short-term improvements in colic and related symptoms in infants with suspected CMPA. These preliminary findings highlight AAF as a promising dietary intervention during early infancy. Larger controlled studies are necessary to confirm these results and explore long-term outcomes.
Collapse
Affiliation(s)
- Jerry M. Brown
- Office of Medical Education, Florida Atlantic University Charles E. Schmidt College of Medicine, Boca Raton, FL 33431, USA; (J.M.B.); (J.V.B.); (L.L.); (J.B.); (J.F.)
| | - Jessica V. Baran
- Office of Medical Education, Florida Atlantic University Charles E. Schmidt College of Medicine, Boca Raton, FL 33431, USA; (J.M.B.); (J.V.B.); (L.L.); (J.B.); (J.F.)
| | - Luke Lamos
- Office of Medical Education, Florida Atlantic University Charles E. Schmidt College of Medicine, Boca Raton, FL 33431, USA; (J.M.B.); (J.V.B.); (L.L.); (J.B.); (J.F.)
| | - Jesse Beacker
- Office of Medical Education, Florida Atlantic University Charles E. Schmidt College of Medicine, Boca Raton, FL 33431, USA; (J.M.B.); (J.V.B.); (L.L.); (J.B.); (J.F.)
| | - Jared Florio
- Office of Medical Education, Florida Atlantic University Charles E. Schmidt College of Medicine, Boca Raton, FL 33431, USA; (J.M.B.); (J.V.B.); (L.L.); (J.B.); (J.F.)
| | - Lea V. Oliveros
- Office of Medical Education, Alabama College of Osteopathic Medicine, Dothan, AL 36303, USA;
| | - Abigail L. Fabbrini
- Office of Medical Education, Kansas City University College of Osteopathic Medicine, Kansas City, MO 64106, USA; (A.L.F.); (A.A.F.)
| | - Andrew A. Farrar
- Office of Medical Education, Kansas City University College of Osteopathic Medicine, Kansas City, MO 64106, USA; (A.L.F.); (A.A.F.)
| | - Michael J. Wilsey
- Department of Pediatrics, University of South Florida Morsani College of Medicine, Tampa, FL 33606, USA
| |
Collapse
|
5
|
Zhu D, Li S, Xu Z, Kulyar MF, Bai X, Wang Y, Wang B, Khateeb E, Deng D, Wang L, Chen Y, Guo A, Shen Y. Comparative analysis of gut microbiota in healthy and diarrheic foals. Microbiol Spectr 2025; 13:e0087124. [PMID: 40105330 PMCID: PMC12054031 DOI: 10.1128/spectrum.00871-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 02/01/2025] [Indexed: 03/20/2025] Open
Abstract
Diarrhea presents a substantial risk of high morbidity and mortality among foals. Although studies have shown connections between gut microbiota and several gastrointestinal diseases, there is still inadequate information on gut microbial alterations in foals during diarrhea. In this study, we conducted 16S rRNA and ITS gene amplicon sequencing to investigate gut bacterial and fungal differences between healthy and diarrheic foals. The results unveiled significant reductions in gut bacterial and fungal diversities among foals experiencing diarrhea, accompanied by notable shifts in the composition of gut microbial communities. A considerable decrease was observed in the relative abundance of 30 bacterial and 34 fungal genera. Moreover, two bacterial and eight fungal genera were utterly undetectable in the gut microbiota of diarrheic foals. Some decreased genera, such as Bifidobacterium and Saccharomyces, were deemed beneficial and recognized as probiotics. The study revealed significant alterations in foals' gut bacterial and fungal communities during diarrhea, which enriched our comprehension of gut microbial dynamics in foals across varying health statuses. These findings offer valuable insights for managing diarrhea through gut microbiota modulation, suggesting that probiotics may be superior to antibiotics in preventing and controlling foal diarrhea.IMPORTANCEThis research advances the understanding of gut bacterial and fungal dynamics in foals, highlighting gut microbiota dysbiosis as a potential contributor to foal diarrhea. Additionally, we observed that many altered bacteria and fungi were downregulated during diarrhea, including some probiotic strains. Consequently, our findings provide evidence that probiotics may offer superior efficacy compared with antibiotics as potential candidates for preventing and treating foal diarrhea.
Collapse
Affiliation(s)
- Di Zhu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Siyu Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zhixiang Xu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Md. F. Kulyar
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xu Bai
- China Horse Industry Association, Beijing, China
| | - Yu Wang
- China Horse Industry Association, Beijing, China
| | - Boya Wang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Emaan Khateeb
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Dandan Deng
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Lidan Wang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yuji Chen
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Aizhen Guo
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yaoqin Shen
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
6
|
Zimmermann P, Kurth S, Giannoukos S, Stocker M, Bokulich NA. NapBiome trial: Targeting gut microbiota to improve sleep rhythm and developmental and behavioural outcomes in early childhood in a birth cohort in Switzerland - a study protocol. BMJ Open 2025; 15:e092938. [PMID: 40032396 PMCID: PMC11877202 DOI: 10.1136/bmjopen-2024-092938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 02/08/2025] [Indexed: 03/05/2025] Open
Abstract
INTRODUCTION The gut-brain axis plays a crucial role in the regulation and development of psychological and physical processes. The first year of life is a critical period for the development of the gut microbiome, which parallels important milestones in establishing sleep rhythm and brain development. Growing evidence suggests that the gut microbiome influences sleep, cognition and early neurodevelopment. For term-born and preterm-born infants, difficulties in sleep regulation may have consequences on health. Identifying effective interventions on the gut-brain axis in early life is likely to have long-term implications for the health and development of at-risk infants. METHODS AND ANALYSES In this multicentre, four-group, double-blinded, placebo (PLC)-controlled randomised trial with a factorial design, 120 preterm-born and 260 term-born infants will be included. The study will investigate whether the administration of daily synbiotics or PLC for a duration of 3 months improves sleep patterns and neurodevelopmental outcomes up to 2 years of age. The trial will also: (1) determine the association between gut microbiota, sleep patterns and health outcomes in children up to 2 years of age; and (2) leverage the interactions between gut microbiota, brain and sleep to develop new intervention strategies for at-risk infants. ETHICS AND DISSEMINATION The NapBiome trial has received ethical approval by the Committee of Northwestern and Central Switzerland and Canton Vaud, Switzerland (#2024-01681). Outcomes will be disseminated through publication and will be presented at scientific conferences. Metagenomic data will be shared through the European Nucleotide Archive. TRIAL REGISTRATION NUMBER The US National Institutes of Health NCT06396689.
Collapse
Affiliation(s)
- Petra Zimmermann
- Department of Community Health and Department of Paediatrics, Fribourg Hospital, University of Fribourg, Fribourg, Switzerland
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Salome Kurth
- Department of Psychology, University of Fribourg, Fribourg, Switzerland
| | - Stamatios Giannoukos
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Martin Stocker
- Neonatology, Children's Hospital Lucerne, Lucerne, Switzerland
| | - Nicholas A Bokulich
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
7
|
Zhang Y, Xu Y, Hu L, Wang X. Advancements related to probiotics for preventing and treating recurrent respiratory tract infections in children. Front Pediatr 2025; 13:1508613. [PMID: 39981209 PMCID: PMC11839809 DOI: 10.3389/fped.2025.1508613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/17/2025] [Indexed: 02/22/2025] Open
Abstract
Recurrent respiratory tract infections (RRTIs) are a common condition in pediatrics and significantly impact children's quality of life; however, their pathogenesis and contributing factors are not yet fully elucidated. Probiotics have recently emerged as promising agents for modulating intestinal microecology and have gained considerable attention in clinical research on preventing and treating RRTIs in children. This article provides an initial overview of the concept, classification, and mechanisms underlying probiotics. It emphasizes their beneficial effects on respiratory health by modulating intestinal microbial equilibrium, augmenting immune system functionality, and attenuating inflammatory responses. Subsequently, we examine existing research regarding the use of probiotics in pediatric RRTIs. Numerous clinical trials have unequivocally demonstrated that supplementing with probiotics can significantly reduce both the frequency and severity of RRTIs in children while also simultaneously decreasing antibiotic usage. However, there are ongoing controversies and challenges in current research concerning the influence of probiotic type, dosage, duration of use, and other factors on efficacy. Furthermore, variations have been observed across different studies. Additionally, it is crucial to further evaluate the safety and potential long-term side effects associated with probiotic use in children with RRTIs. In conclusion, we propose future research directions including conducting more high-quality randomized controlled trials to optimize application strategies for probiotics alongside other treatments while considering variations based on age and health conditions among pediatric populations. Finally, in summary although probiotics exhibit promising benefits in preventing and treating RRTIs in children; additional studies are necessary to refine their application strategies ensuring both safety and effectiveness.
Collapse
Affiliation(s)
- Yali Zhang
- Tianyou Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Yingying Xu
- Tianyou Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Ling Hu
- Tianyou Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Xiaomei Wang
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
8
|
Abd El-Salam MH, El-Shibiny S, Assem FM, El-Sayyad GS, Hasanien YA, Elfadil D, Soliman TN. Impact of Fermented Milk On Gut Microbiota And Human Health: A Comprehensive Review. Curr Microbiol 2025; 82:107. [PMID: 39888432 DOI: 10.1007/s00284-025-04061-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 01/02/2025] [Indexed: 02/01/2025]
Abstract
The beneficial impact of gut microbiota on human health has encouraged studies on factors modulating it. Among the different factors, diet plays a vital role in this area. Many studies on animals and humans have been concerned with the effects of fermented milk products on gut microbiota and how they relate to health benefits. Yoghurt, kefir, Koumiss, and fermented kinds of milk made using different probiotic strains were tested for their capability to modulate gut microbiota. It is apparent that the microflora present in fermented milk, specifically probiotics, are capable of enduring the gastrointestinal tract's adverse conditions primarily through transit microorganisms. Meanwhile, they can alter the gut microbiota in several ways that benefit human health. The present article gives a comprehensive overview of the modulation of gut microbiota by consumption of fermented milk, particularly those containing probiotics, and their impact on human health.
Collapse
Affiliation(s)
| | | | | | - Gharieb S El-Sayyad
- Medical Laboratory Technology Department, Faculty of Applied Health Sciences Technology, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt.
- Drug Microbiology Laboratory, Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt.
| | - Yasmeen A Hasanien
- Microbiology Laboratory, Plant Research Department, Nuclear Research Center, Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| | - Dounia Elfadil
- Biology and Chemistry Department, Faculty of Sciences and Techniques, Hassan II University of Casablanca, Casablanca, Morocco
| | | |
Collapse
|
9
|
Delcourt H, Huysentruyt K, Vandenplas Y. A synbiotic mixture for the management of infantile colic: A randomized trial. Eur J Pediatr 2024; 184:27. [PMID: 39557731 PMCID: PMC11573799 DOI: 10.1007/s00431-024-05860-5] [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: 06/12/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 11/20/2024]
Abstract
Infant colic is defined as a recurrent and prolonged period of fussing, crying and/or irritability that cannot be prevented or resolved by caregivers. The aim of this study is to evaluate the efficacy of a synbiotic (Bactecal D Liquid) in infants consulting a primary health care professional for inconsolable crying. A randomized trial was conducted in 68 infants diagnosed by the consulted primary health care professional as "probably suffering from infant colic". Patients were randomized into two groups and given the synbiotic once (group 1) or twice (group 2) a day for 28 days. Quality of life (QoL) of the caregivers, evaluated with a Likert scale, was the primary outcome. Secondary outcomes included the total number of crying episodes, total crying time, gassiness and "balling of the fists". The median (Q1;Q3) QoL scores were significantly (p < 0.001) higher on day 28 than at baseline: 6 (5;7) vs 2 (1;3). At baseline, there was no significant difference (p = 0.527) in QoL between both groups. The improvement in QoL was already significant after one week of intervention for both groups. The median number of crying episodes, overall crying time, gassiness and "balling of fists" were significantly lower on day 28 compared to baseline (p < 0.001). CONCLUSION The synbiotic tested was shown to be efficacious in the management of infant colic. A significant improvement was observed after 7 days of intervention, which is much earlier than the expected decrease related to the natural evolution of infant colic. WHAT IS KNOWN • Some probiotic strains are reported to be effective in the management of infants presenting with colic, if breastfed. WHAT IS NEW • The synbiotic studied improved quality of life of caregivers of infants presenting infant colic. • Two doses of the synbiotic were not more effective than one dose. • The improved occurred within one week. • The improvement was independent of feeding (breastfeeding, formula feeding or mixed feeding).
Collapse
Affiliation(s)
- Hanne Delcourt
- Vrije Universiteit Brussel (VUB), UZ Brussels, KidZ Health Castle, Laarbeeklaan 101, 1090, Brussels, Belgium
| | - Koen Huysentruyt
- Vrije Universiteit Brussel (VUB), UZ Brussels, KidZ Health Castle, Laarbeeklaan 101, 1090, Brussels, Belgium
| | - Yvan Vandenplas
- Vrije Universiteit Brussel (VUB), UZ Brussels, KidZ Health Castle, Laarbeeklaan 101, 1090, Brussels, Belgium.
| |
Collapse
|
10
|
Guamán LP, Carrera-Pacheco SE, Zúñiga-Miranda J, Teran E, Erazo C, Barba-Ostria C. The Impact of Bioactive Molecules from Probiotics on Child Health: A Comprehensive Review. Nutrients 2024; 16:3706. [PMID: 39519539 PMCID: PMC11547800 DOI: 10.3390/nu16213706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024] Open
Abstract
Background: This review investigates the impact of bioactive molecules produced by probiotics on child health, focusing on their roles in modulating gut microbiota, enhancing immune function, and supporting overall development. Key metabolites, including short-chain fatty acids (SCFAs), bacteriocins, exopolysaccharides (EPSs), vitamins, and gamma-aminobutyric acid (GABA), are highlighted for their ability to maintain gut health, regulate inflammation, and support neurodevelopment. Objectives: The aim of this review is to examine the mechanisms of action and clinical evidence supporting the use of probiotics and postbiotics in pediatric healthcare, with a focus on promoting optimal growth, development, and overall health in children. Methods: The review synthesizes findings from clinical studies that investigate the effects of probiotics and their metabolites on pediatric health. The focus is on specific probiotics and their ability to influence gut health, immune responses, and developmental outcomes. Results: Clinical studies demonstrate that specific probiotics and their metabolites can reduce gastrointestinal disorders, enhance immune responses, and decrease the incidence of allergies and respiratory infections in pediatric populations. Additionally, postbiotics-bioactive compounds from probiotic fermentation-offer promising benefits, such as improved gut barrier function, reduced inflammation, and enhanced nutrient absorption, while presenting fewer safety concerns compared to live probiotics. Conclusions: By examining the mechanisms of action and clinical evidence, this review underscores the potential of integrating probiotics and postbiotics into pediatric healthcare strategies to promote optimal growth, development, and overall health in children.
Collapse
Affiliation(s)
- Linda P. Guamán
- Centro de Investigación Biomédica (CENBIO), Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170527, Ecuador; (S.E.C.-P.); (J.Z.-M.)
| | - Saskya E. Carrera-Pacheco
- Centro de Investigación Biomédica (CENBIO), Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170527, Ecuador; (S.E.C.-P.); (J.Z.-M.)
| | - Johana Zúñiga-Miranda
- Centro de Investigación Biomédica (CENBIO), Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170527, Ecuador; (S.E.C.-P.); (J.Z.-M.)
| | - Enrique Teran
- Colegio de Ciencias de la Salud, Universidad San Francisco de Quito USFQ, Quito 170901, Ecuador; (E.T.); (C.E.)
| | - Cesar Erazo
- Colegio de Ciencias de la Salud, Universidad San Francisco de Quito USFQ, Quito 170901, Ecuador; (E.T.); (C.E.)
| | - Carlos Barba-Ostria
- Colegio de Ciencias de la Salud, Universidad San Francisco de Quito USFQ, Quito 170901, Ecuador; (E.T.); (C.E.)
- Instituto de Microbiología, Universidad San Francisco de Quito USFQ, Quito 170901, Ecuador
| |
Collapse
|
11
|
Whitmore M, Tobin I, Burkardt A, Zhang G. Nutritional Modulation of Host Defense Peptide Synthesis: A Novel Host-Directed Antimicrobial Therapeutic Strategy? Adv Nutr 2024; 15:100277. [PMID: 39053604 PMCID: PMC11381887 DOI: 10.1016/j.advnut.2024.100277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/11/2024] [Accepted: 07/19/2024] [Indexed: 07/27/2024] Open
Abstract
The escalating threat of antimicrobial resistance underscores the imperative for innovative therapeutic strategies. Host defense peptides (HDPs), integral components of innate immunity, exhibit profound antimicrobial and immunomodulatory properties. Various dietary compounds, such as short-chain fatty acids, vitamins, minerals, sugars, amino acids, phytochemicals, bile acids, probiotics, and prebiotics have been identified to enhance the synthesis of endogenous HDPs without provoking inflammatory response or compromising barrier integrity. Additionally, different classes of these compounds synergize in augmenting HDP synthesis and disease resistance. Moreover, dietary supplementation of several HDP-inducing compounds or their combinations have demonstrated robust protection in rodents, rabbits, pigs, cattle, and chickens from experimental infections. However, the efficacy of these compounds in inducing HDP synthesis varies considerably among distinct compounds. Additionally, the regulation of HDP genes occurs in a gene-specific, cell type-specific, and species-specific manner. In this comprehensive review, we systematically summarized the modulation of HDP synthesis and the mechanism of action attributed to each major class of dietary compounds, including their synergistic combinations, across a spectrum of animal species including humans. We argue that the ability to enhance innate immunity and barrier function without triggering inflammation or microbial resistance positions the nutritional modulation of endogenous HDP synthesis as a promising host-directed approach for mitigating infectious diseases and antimicrobial resistance. These HDP-inducing compounds, particularly in combinations, harbor substantial clinical potential for further exploration in antimicrobial therapies for both human and other animals.
Collapse
Affiliation(s)
- Melanie Whitmore
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK, United States
| | - Isabel Tobin
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK, United States
| | - Amanda Burkardt
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK, United States
| | - Guolong Zhang
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK, United States.
| |
Collapse
|
12
|
Sun Y, Guo S, Kwok LY, Sun Z, Wang J, Zhang H. Probiotic Bifidobacterium animalis ssp. lactis Probio-M8 improves the fermentation and probiotic properties of fermented milk. J Dairy Sci 2024; 107:6643-6657. [PMID: 38825144 DOI: 10.3168/jds.2024-24863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 04/08/2024] [Indexed: 06/04/2024]
Abstract
Probiotics are increasingly used as starter cultures to produce fermented dairy products; however, few studies have investigated the role of probiotics in milk fermentation metabolism. The current study aimed to investigate whether adding Bifidobacterium animalis ssp. lactis Probio-M8 (Probio-M8) as a starter culture strain could improve milk fermentation by comparing the physicochemical characteristics and metabolomes of fermented milks produced by a commercial starter culture with and without Probio-M8. Our results showed that adding Probio-M8 shortened the milk fermentation time and improved the fermented milk texture and stability. Metabolomics analyses revealed that adding Probio-M8 affected mostly organic acid, AA, and fatty acid metabolism in milk fermentation. Targeted quantitative analyses revealed significant increases in various metabolites related to the sensory quality, nutritive value, and health benefits of the probiotic fermented milk, including 5 organic acids (acetic acid, lactic acid, citric acid, succinic acid, and tartaric acid), 5 EAA (valine, arginine, leucine, isoleucine, and lysine), glutamic acid, and 2 essential fatty acids (α-linolenic acid and docosahexaenoic acid). Thus, applying probiotics in milk fermentation is desirable. This study has generated useful information for developing novel functional dairy products.
Collapse
Affiliation(s)
- Yaru Sun
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, 010018, PR China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, 010018, PR China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, PR China
| | - Shuai Guo
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, 010018, PR China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, 010018, PR China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, PR China
| | - Lai-Yu Kwok
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, 010018, PR China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, 010018, PR China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, PR China
| | - Zhihong Sun
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, 010018, PR China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, 010018, PR China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, PR China
| | - Jicheng Wang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, 010018, PR China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, 010018, PR China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, PR China
| | - Heping Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, 010018, PR China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, 010018, PR China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, PR China.
| |
Collapse
|
13
|
Ermann Lundberg L, Pallabi Mishra P, Liu P, Forsberg MM, Sverremark-Ekström E, Grompone G, Håkansson S, Linninge C, Roos S. Bifidobacterium longum subsp. longum BG-L47 boosts growth and activity of Limosilactobacillus reuteri DSM 17938 and its extracellular membrane vesicles. Appl Environ Microbiol 2024; 90:e0024724. [PMID: 38888338 PMCID: PMC11267924 DOI: 10.1128/aem.00247-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/14/2024] [Indexed: 06/20/2024] Open
Abstract
The aim of this study was to identify a Bifidobacterium strain that improves the performance of Limosilactobacillus reuteri DSM 17938. Initial tests showed that Bifidobacterium longum subsp. longum strains boosted the growth of DSM 17938 during in vivo-like conditions. Further characterization revealed that one of the strains, BG-L47, had better bile and acid tolerance compared to BG-L48, as well as mucus adhesion compared to both BG-L48 and the control strain BB536. BG-L47 also had the capacity to metabolize a broad range of carbohydrates and sugar alcohols. Mapping of glycoside hydrolase (GH) genes of BG-L47 and BB536 revealed many GHs associated with plant-fiber utilization. However, BG-L47 had a broader phenotypic fiber utilization capacity. In addition, B. longum subsp. longum cells boosted the bioactivity of extracellular membrane vesicles (MV) produced by L. reuteri DSM 17938 during co-cultivation. Secreted 5'-nucleotidase (5'NT), an enzyme that converts AMP into the signal molecule adenosine, was increased in MV boosted by BG-L47. The MV exerted an improved antagonistic effect on the pain receptor transient receptor potential vanilloid 1 (TRPV1) and increased the expression of the immune development markers IL-6 and IL-1ß in a peripheral blood mononuclear cell (PBMC) model. Finally, the safety of BG-L47 was evaluated both by genome safety assessment and in a human safety study. Microbiota analysis showed that the treatment did not induce significant changes in the composition. In conclusion, B. longum subsp. longum BG-L47 has favorable physiological properties, can boost the in vitro activity of L. reuteri DSM 17938, and is safe for consumption, making it a candidate for further evaluation in probiotic studies. IMPORTANCE By using probiotics that contain a combination of strains with synergistic properties, the likelihood of achieving beneficial interactions with the host can increase. In this study, we first performed a broad screening of Bifidobacterium longum subsp. longum strains in terms of synergistic potential and physiological properties. We identified a superior strain, BG-L47, with favorable characteristics and potential to boost the activity of the known probiotic strain Limosilactobacillus reuteri DSM 17938. Furthermore, we demonstrated that BG-L47 is safe for consumption in a human randomized clinical study and by performing a genome safety assessment. This work illustrates that bacteria-bacteria interactions differ at the strain level and further provides a strategy for finding and selecting companion strains of probiotics.
Collapse
Affiliation(s)
- Ludwig Ermann Lundberg
- Department of Molecular Sciences, Uppsala BioCenter, Swedish University of Agricultural Sciences, Uppsala, Sweden
- BioGaia, Stockholm, Sweden
| | - Punya Pallabi Mishra
- Department of Molecular Sciences, Uppsala BioCenter, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | | | - Manuel Mata Forsberg
- The Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Eva Sverremark-Ekström
- The Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | | | - Sebastian Håkansson
- BioGaia, Stockholm, Sweden
- Division of Applied Microbiology, Department of Chemistry, Lund University, Lund, Sweden
| | - Caroline Linninge
- BioGaia, Stockholm, Sweden
- Department of Food Technology, Engineering and Nutrition, Lund University, Lund, Sweden
| | - Stefan Roos
- Department of Molecular Sciences, Uppsala BioCenter, Swedish University of Agricultural Sciences, Uppsala, Sweden
- BioGaia, Stockholm, Sweden
| |
Collapse
|
14
|
Lan W, Yang H, Zhong Z, Luo C, Huang Q, Liu W, Yang J, Xiang H, Tang Y, Chen T. Bifidobacterium animalis subsp. lactis LPL-RH improves postoperative gastrointestinal symptoms and nutrition indexes by regulating the gut microbiota in patients with valvular heart disease: a randomized controlled trial. Food Funct 2024; 15:7605-7618. [PMID: 38938120 DOI: 10.1039/d4fo01471e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
Gastrointestinal symptoms constitute a frequent complication in postoperative patients with valvular heart disease (VHD), impacting their postoperative recovery. Probiotics contribute to regulating human gut microbiota balance and alleviating postoperative gastrointestinal symptoms. Our objective involved assessing the potential of Bifidobacterium animalis subsp. lactis LPL-RH to alleviate postoperative gastrointestinal symptoms and expedite patient recovery. Adult patients diagnosed with VHD scheduled for valve surgery were enrolled. 110 patients were randomly divided into two groups and received LPL-RH or a placebo for 14 days. Gastrointestinal symptoms were evaluated using the Gastrointestinal Symptoms Questionnaire. An analysis of the time to recovery of bowel function and various postoperative variables was conducted in both study groups. Variations in the intestinal microbiota were detected via 16S rRNA sequencing. The study was completed by 105 participants, with 53 in the probiotic group and 52 in the placebo group. Compared to the placebo group, LPL-RH significantly reduced the total gastrointestinal symptom score after surgery (p = 0.004). Additionally, LPL-RH was found to significantly reduce abdominal pain (p = 0.001), bloating (p = 0.018), and constipation (p = 0.022) symptom scores. Furthermore, LPL-RH dramatically shortened the time to recovery of bowel function (p = 0.017). Moreover, LPL-RH administration significantly enhanced patients' postoperative nutrition indexes (red blood cell counts, hemoglobin level, p < 0.05). Microbiome analysis showed that the composition and diversity of the postoperative intestinal microbiota differed between the probiotic and placebo groups. No adverse incidents associated with probiotics were documented, emphasizing their safety. This study initially discovered that oral B. animalis subsp. lactis LPL-RH can assist in regulating intestinal microbiota balance, alleviating gastrointestinal symptoms, promoting intestinal function recovery, and enhancing nutrition indexes in patients with VHD after surgery. Regulating the intestinal microbiota may represent a potential mechanism for LPL-RH to exert clinical benefits.
Collapse
Affiliation(s)
- Wanqi Lan
- Department of Cardiovascular Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- The Second Clinical Medical College, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Heng Yang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- The Second Clinical Medical College, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zhiwang Zhong
- Department of Cardiovascular Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- The Second Clinical Medical College, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Chao Luo
- Department of Cardiovascular Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- The Second Clinical Medical College, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Qin Huang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- The Second Clinical Medical College, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Wu Liu
- Department of Cardiovascular Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- The Second Clinical Medical College, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Juesheng Yang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| | - Haiyan Xiang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| | - Yanhua Tang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| | - Tingtao Chen
- Department of Cardiovascular Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- The Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, China
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
15
|
Guarner F, Sanders ME, Szajewska H, Cohen H, Eliakim R, Herrera-deGuise C, Karakan T, Merenstein D, Piscoya A, Ramakrishna B, Salminen S, Melberg J. World Gastroenterology Organisation Global Guidelines: Probiotics and Prebiotics. J Clin Gastroenterol 2024; 58:533-553. [PMID: 38885083 DOI: 10.1097/mcg.0000000000002002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 03/03/2024] [Indexed: 06/20/2024]
Affiliation(s)
| | - Mary Ellen Sanders
- International Scientific Association for Probiotics and Prebiotics, Centennial, CO
| | - Hania Szajewska
- Department of Paediatrics, The Medical University of Warsaw, Warsaw, Poland
| | | | | | | | | | | | | | | | | | - Jim Melberg
- World Gastroenterology Organisation, Milwaukee, WI
| |
Collapse
|
16
|
Chen K, Jin S, Ma Y, Cai L, Xu P, Nie Y, Luo L, Yu Q, Shen Y, Zhou Z, Liu C. Adjudicative efficacy of Bifidobacterium animalis subsp. lactis BLa80 in treating acute diarrhea in children: a randomized, double-blinded, placebo-controlled study. Eur J Clin Nutr 2024; 78:501-508. [PMID: 38467857 PMCID: PMC11182741 DOI: 10.1038/s41430-024-01428-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 02/28/2024] [Accepted: 03/01/2024] [Indexed: 03/13/2024]
Abstract
The goal of this study is to assess the efficacy and safety of Bifidobacterium animalis subsp. lactis BLa80, as an adjunct treatment for diarrhea in children with a randomized, double-blinded, placebo-controlled study design. Eligible diarrheal children, aged 0-3 years without the need for antibiotic treatment based on clinical diagnosis when recruited, were randomized into the intervention group (IG, n = 58, with probiotic) or the control group (CG, n = 53, placebo). The primary assessment was the duration of diarrhea. Fecal samples were collected for biochemical index measurement, analysis of gut microbiome composition, and prediction of gene family abundances. The total duration of diarrhea in the IG (122.6 ± 13.1 h) was significantly shorter than in the CG (148.4 ± 17.6 h, p < 0.001). More children in the IG showed improvements in diarrhea compared to the CG, both in intention-to-treat analysis (81.7% vs. 40.0%, p < 0.001) and per protocol analysis (84.4% vs 45.3%, p < 0.001). Cathelicidin level in the IG was significantly higher than that in the CG after the intervention (4415.00 ± 1036.93 pg/g vs. 3679.49 ± 871.18 pg/g, p = 0.0175). The intervention led to an increased abundance of Bifidobacterium breve and Collinsella aerofaciens species, higher alpha-diversity (p < 0.05), and enrichment of functional genes in the gut microbiota related to immunity regulation. Administration of BLa80 at a dose of 5 × 109 CFU/day resulted in a shorter duration of diarrhea and alterations in gut microbiome composition and gene functions.
Collapse
Affiliation(s)
- Ke Chen
- Department of Nutrition, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| | - Shanshan Jin
- Department of Nutrition, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yu Ma
- Department of Neonatology, Dayi Maternal and Child Health Care Hospital, Chengdu, China
| | - Limei Cai
- Department of Neonatology, Qingbaijiang Maternal and Child Health Care Hospital, Chengdu, China
| | - Ping Xu
- Department of Child Health Care, Qingbaijiang Maternal and Child Health Care Hospital, Chengdu, China
| | - Yang Nie
- Department of Child Health Care, Chongzhou Maternal and Child Health Care Hospital, Chengdu, China
| | - Li Luo
- Department of Pediatrics, Dayi Maternal and Child Health Care Hospital, Chengdu, China
| | - Qinghua Yu
- Laboratory of Microbiology, Immunology and Metabolism, Diprobio (Shanghai) Co., Limited, Shanghai, China
| | - Yang Shen
- Laboratory of Microbiology, Immunology and Metabolism, Diprobio (Shanghai) Co., Limited, Shanghai, China
| | - Zengyuan Zhou
- Department of Nutrition, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Changqi Liu
- School of Exercise and Nutritional Sciences, San Diego State University, San Diego, CA, USA
| |
Collapse
|
17
|
Chen K, Zhou Z, Nie Y, Cao Y, Yang P, Zhang Y, Xu P, Yu Q, Shen Y, Ma W, Jin S, Liu C. Adjunctive efficacy of Bifidobacterium animalis subsp. lactis XLTG11 for functional constipation in children. Braz J Microbiol 2024; 55:1317-1330. [PMID: 38381349 PMCID: PMC11153453 DOI: 10.1007/s42770-024-01276-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 02/07/2024] [Indexed: 02/22/2024] Open
Abstract
Functional constipation (FC) can seriously affect the physical and mental health of children. The goal of this study is to assess the efficacy and safety of Bifidobacterium animalis subsp. lactis XLTG11 in treating FC in children through a randomized, double-blinded, placebo-controlled approach. Eligible children were randomized into either the intervention group (IG, n = 65, receiving conventional treatment with probiotics) or the control group (CG, n = 66, receiving conventional treatment without probiotics). The primary outcome measure was fecal frequency. Fecal gut microbiota analysis and PICRUSt (Phylogenetic Investigation of Communities by Reconstruction of Unobserved States) were used to predict gene family abundances based on 16S information. Over the course of treatment, the weekly frequency of feces within each group increased significantly (F = 41.97, p < 0.001). The frequency of feces (times/week (t/w)) in the IG was significantly higher than that in the CG (3.69 ± 2.62 t/w vs.3.18 ± 1.43 t/w, 4.03 ± 2.54 t/w vs. 2.89 ± 1.39 t/w and 3.74 ± 2.36 t/w vs. 2.94 ± 1.18 t/w and 3.45 ± 1.98 vs. 3.17 ± 1.41 t/w for the 1st, 2nd, 3rd, and 4th week after intervention, respectively) (F = 7.60, p = 0.0067). After the intervention, dominate species shifted to Bifidobacterium longum, Bifidobacterium breve, and Escherichia coli in the IG. Additionally, genes related to short-chain fatty acid (SCF) metabolism were upregulated, while methane metabolism was downregulated. Administration of XLTG11 at a dose of 1 × 1010 CFU/day to children increased fecal frequency, induced beneficial changes in gut microbiota, and regulated SCFs and methane metabolism-related genes.
Collapse
Affiliation(s)
- Ke Chen
- Department of Nutrition, School of Medicine, Chengdu Women's & Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| | - Zengyuan Zhou
- Department of Nutrition, School of Medicine, Chengdu Women's & Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yang Nie
- Department of Child Health Care, Chongzhou Maternal and Child Health Care Hospital, Chengdu, China
| | - Yanmei Cao
- Department of Child Health Care, Dayi Maternal and Child Health Care Hospital, Chengdu, China
| | - Ping Yang
- Department of Child Health Care, Xindu Maternal and Child Health Care Hospital, Chengdu, China
| | - Ying Zhang
- Department of Child Health Care, Jinniu Maternal and Child Health Care Hospital, Chengdu, China
| | - Ping Xu
- Department of Child Health Care, Qingbaijiang Maternal and Child Health Care Hospital, Chengdu, China
| | - Qinghua Yu
- Laboratory of Microbiology, Immunology and Metabolism, Diprobio (Shanghai) Co, Limited, Shanghai, China
| | - Yang Shen
- Laboratory of Microbiology, Immunology and Metabolism, Diprobio (Shanghai) Co, Limited, Shanghai, China
| | - Weiwei Ma
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shanshan Jin
- Department of Nutrition, School of Medicine, Chengdu Women's & Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Changqi Liu
- School of Exercise and Nutritional Sciences, San Diego State University, San Diego, USA
| |
Collapse
|
18
|
Güler MA, Çetin B, Albayrak B, Meral-Aktaş H, Tekgündüz KŞ, Kara M, Işlek A. Isolation, identification, and in vitro probiotic characterization of forty novel Bifidobacterium strains from neonatal feces in Erzurum province, Türkiye. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:4165-4175. [PMID: 38299445 DOI: 10.1002/jsfa.13298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 02/02/2024]
Abstract
BACKGROUND Neonatal feces are one of the most important sources for probiotic isolation. The purpose of this study was the isolation and identification of Bifidobacterium spp. from neonatal feces and the evaluation of in vitro probiotic properties of strains including safety tests. RESULTS A total of 40 isolates were obtained from 14 healthy newborns' feces in Erzurum province, Türkiye. By their rep-PCR patterns and 16S rRNA gene sequences, isolates were identified as 26 Bifidobacterium breve and 14 Bifidobacterium longum. Fifteen of the isolates tolerated bile salts and showed high resistance to simulated gastric juice. Isolates exhibited varying rates of auto-aggregation and hydrophobicity. In addition, most of the isolates displayed antibacterial activity against Escherichia coli O157:H7, Staphylococcus aureus ATCC 29213, Salmonella Typhimurium RSHMB 95091, and Pseudomonas aeruginosa ATCC 9027. However, only one strain showed bile salt hydrolase activity and two strains showed the ability to produce H2O2. Bifidobacterium strains were generally sensitive to the tested antibiotics and lacked kanamycin, gentamicin, and streptomycin resistance genes, and hemolytic and DNAse activities. On the other hand, it was determined that five strains had various virulence genes including gelE, esp, efaAfs, hyl, and ace. CONCLUSION Results of the present study suggested that B. longum BH28, B. breve BH4 and B. breve BH5 strains have the potential as probiotic candidates for further studies. © 2024 The Authors. Journal of The Science of Food and Agriculture published by John Wiley & Sons Ltd on behalf of Society of Chemical Industry.
Collapse
Affiliation(s)
- Muhammet Akif Güler
- Division of Pediatric Nephrology, Department of Pediatrics, Atatürk University, Faculty of Medicine, Erzurum, Türkiye
| | - Bülent Çetin
- Department of Food Engineering, Faculty of Agriculture, Ataturk University, Erzurum, Türkiye
| | - Bülent Albayrak
- Department of Gastroenterology, Faculty of Medicine, Atatürk University, Erzurum, Türkiye
| | - Hacer Meral-Aktaş
- Department of Food Engineering, Faculty of Agriculture, Ataturk University, Erzurum, Türkiye
| | - Kadir Şerafettin Tekgündüz
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Atatürk University, Erzurum, Türkiye
| | - Mustafa Kara
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Atatürk University, Erzurum, Türkiye
| | - Ali Işlek
- Department of Pediatric Gastroenterology, Faculty of Medicine, Çukurova University, Adana, Türkiye
| |
Collapse
|
19
|
Ding M, Li B, Chen H, Ross RP, Stanton C, Zhao J, Chen W, Yang B. Bifidobacterium longum Subsp. infantis Promotes IgA Level of Growing Mice in a Strain-Specific and Intestinal Niche-Dependent Manner. Nutrients 2024; 16:1148. [PMID: 38674840 PMCID: PMC11054607 DOI: 10.3390/nu16081148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/06/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Throughout infancy, IgA is crucial for maintaining gut mucosal immunity. This study aims to determine whether supplementing newborn mice with eight different strains of Bifidobacterium longum subsp. infantis might regulate their IgA levels. The strains were gavaged to BALB/C female (n = 8) and male (n = 8) dams at 1-3 weeks old. Eight strains of B. longum subsp. infantis had strain-specific effects in the regulation of intestinal mucosal barriers. B6MNI, I4MI, and I10TI can increase the colonic IgA level in females and males. I8TI can increase the colonic IgA level in males. B6MNI was also able to significantly increase the colonic sIgA level in females. B6MNI, I4MI, I8TI, and I10TI regulated colonic and Peyer's patch IgA synthesis genes but had no significant effect on IgA synthesis pathway genes in the jejunum and ileum. Moreover, the variety of sIgA-coated bacteria in male mice was changed by I4MI, I5TI, I8TI, and B6MNI. These strains also can decrease the relative abundance of Escherichia coli. These results indicate that B. longum subsp. infantis can promote IgA levels but show strain specificity. Different dietary habits with different strains of Bifidobacterium may have varying effects on IgA levels when supplemented in early infancy.
Collapse
Affiliation(s)
- Mengfan Ding
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (M.D.); (B.L.); (H.C.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Bowen Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (M.D.); (B.L.); (H.C.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Haiqin Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (M.D.); (B.L.); (H.C.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Reynolds Paul Ross
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi 214122, China; (R.P.R.); (C.S.)
- APC Microbiome Ireland, University College Cork, T12 R229 Cork, Ireland
| | - Catherine Stanton
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi 214122, China; (R.P.R.); (C.S.)
- APC Microbiome Ireland, University College Cork, T12 R229 Cork, Ireland
- Teagasc Food Research Centre, Moorepark, Fermoy, P61 C996 Cork, Ireland
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (M.D.); (B.L.); (H.C.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (M.D.); (B.L.); (H.C.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Bo Yang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (M.D.); (B.L.); (H.C.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi 214122, China; (R.P.R.); (C.S.)
| |
Collapse
|
20
|
Xing M, Li X, Zhang Y. Analysis of Bifidobacterium animalis subsp. lactis BB-12 ® and Lactobacillus rhamnosus GG on underweight and malabsorption in premature infants. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2024; 70:e20230636. [PMID: 38422245 PMCID: PMC10890203 DOI: 10.1590/1806-9282.20230636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 09/24/2023] [Indexed: 03/02/2024]
Abstract
OBJECTIVE This study aimed to explore and analyze the therapeutic effect of the combination of Bifidobacterium animalis subsp. lactis BB-12® and Lactobacillus rhamnosus GG on underweight and malabsorption in premature infants. METHODS This is a retrospective study. The clinical data of 68 premature infants admitted to Beijing United Family Hospital (Private Secondary Comprehensive Hospital, Chaoyang District, Beijing, China) from January 2016 to January 2022 were analyzed retrospectively. Preterm infants less than 37 weeks of gestational age admitted to the neonatal intensive care unit were included in the study. Patients with intestinal malformations, necrotizing enterocolitis, etc., who require long-term fasting were excluded. A telephone follow-up was performed 3-6 months after discharge. They were classified as treatment groups A and B according to the treatment plan. The treatment group A included parenteral nutrition, enteral nutrition, etc. In treatment group B, based on treatment group A, the premature infants were treated with Bifidobacterium animalis subsp. lactis BB-12® and Lactobacillus rhamnosus GG. The time to regain birthweight and the weight on day 30 were compared between the two groups, as was the duration of transition from parenteral nutrition to total enteral nutrition. RESULTS The time of weight regain birthweight in group B was shorter than that in group A (t=-2.560; t=-4.287; p<0.05). The increase of weight on day 30 in group B was significantly higher than that in group A (t=2.591; t=2.651; p<0.05). The time from parenteral nutrition to total enteral nutrition in group B was shorter than that in group A (z=-2.145; z=-2.236; p<0.05). CONCLUSION In the treatment of premature infants, the combination of Bifidobacterium animalis subsp. lactis BB-12® and Lactobacillus rhamnosus GG can have a better therapeutic effect on the underweight and malabsorption of premature infants, and this treatment method can be popularized in clinics.
Collapse
Affiliation(s)
- Meng Xing
- Beijing United Family Hospital, Department of Pediatrics - Beijing, China
| | - Xuran Li
- Beijing United Family Jingbei Women and Children's Hospital, Department of Pediatrics - Beijing, China
| | - Yinzhu Zhang
- Beijing United Family Jingbei Women and Children's Hospital, Department of Pediatrics - Beijing, China
| |
Collapse
|
21
|
Vaz SR, Tofoli MH, Avelino MAG, da Costa PSS. Probiotics for infantile colic: Is there evidence beyond doubt? A meta-analysis and systematic review. Acta Paediatr 2024; 113:170-182. [PMID: 37962097 DOI: 10.1111/apa.17036] [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: 09/09/2023] [Revised: 10/27/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023]
Abstract
AIM This study is a systematic review and meta-analysis of randomised controlled trials that employed probiotics and symbiotics for treating infantile colic. METHODS We performed electronic systematic literature searches in Embase, PubMed and Web of Science, to identify articles published between 1950 and April 2023. Only RCT involving infants with infantile colic under 3 months were included. The treatment plan comprised 15 probiotics, which included Lactobacillus reuteri DSM 17938 and Bifidobacterium animalis lactis BB-12. The probiotics were administered alone or in combination with a prebiotic, vs. no intervention or a placebo. RESULTS Probiotics resulted in an average reduction of 51 min of crying per day (p = 0.001). Further analysis of subgroups showed that the reduction was -39.30 min for vaginal delivery (p = 0.003), -64.66 min for Lactobacillus reuteri DSM 17938 (p = 0.03), -40.45 min for other strains (p < 0.00001), -74.28 min for exclusively breastfed infants (p = 0.0003) and -48.04 min for mixed feeding (p < 0.00001). CONCLUSION All probiotic strains seem effective in treating infantile colic. Exclusively breastfed infants have demonstrated more significant reduction in crying time. However, the available evidence on the effectiveness of probiotics in formula-fed and caesarean-born infants is limited.
Collapse
Affiliation(s)
| | - Marise Helena Tofoli
- Department of Pediatric Gastroenterology, State Hospital for Children and Adolescents, Goiânia, Brazil
| | | | | |
Collapse
|
22
|
Zhou T, Xiao L, Zuo Z, Zhao F. MAMI: a comprehensive database of mother-infant microbiome and probiotic resources. Nucleic Acids Res 2024; 52:D738-D746. [PMID: 37819042 PMCID: PMC10767955 DOI: 10.1093/nar/gkad813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/04/2023] [Accepted: 09/19/2023] [Indexed: 10/13/2023] Open
Abstract
Extensive evidence has demonstrated that the human microbiome and probiotics confer great impacts on human health, particularly during critical developmental stages such as pregnancy and infancy when microbial communities undergo remarkable changes and maturation. However, a major challenge in understanding the microbial community structure and interactions between mothers and infants lies in the current lack of comprehensive microbiome databases specifically focused on maternal and infant health. To address this gap, we have developed an extensive database called MAMI (Microbiome Atlas of Mothers and Infants) that archives data on the maternal and neonatal microbiome, as well as abundant resources on edible probiotic strains. By leveraging this resource, we can gain profound insights into the dynamics of microbial communities, contributing to lifelong wellness for both mothers and infants through precise modulation of the developing microbiota. The functionalities incorporated into MAMI provide a unique perspective on the study of the mother-infant microbiome, which not only advance microbiome-based scientific research but also enhance clinical practice. MAMI is publicly available at https://bioinfo.biols.ac.cn/mami/.
Collapse
Affiliation(s)
- Tian Zhou
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Liwen Xiao
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhenqiang Zuo
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China
| | - Fangqing Zhao
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
23
|
Lin C, Lin Y, Wang S, Wang J, Mao X, Zhou Y, Zhang H, Chen W, Wang G. Bifidobacterium animalis subsp. lactis boosts neonatal immunity: unravelling systemic defences against Salmonella. Food Funct 2024; 15:236-254. [PMID: 38054827 DOI: 10.1039/d3fo03686c] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
Bifidobacterium animalis subsp. lactis may be a useful probiotic intervention for regulating neonatal intestinal immune responses and counteracting Salmonella infection. However, recent research has focused on intestinal immunity, leaving uncertainties regarding the central, peripheral, and neural immune responses in neonates. Therefore, this study investigated the role and mechanisms of B. animalis subsp. lactis in the systemic immune responses of neonatal rats following Salmonella infection. Through extremely early pretreatment with B. animalis subsp. lactis (6 hours postnatal), the neonatal rat gut microbiota was effectively reshaped, especially the Bifidobacterium community. In the rats pretreated with B. animalis subsp. lactis, Salmonella was less prevalent in the blood, liver, spleen, and intestines following infection. The intervention promoted T lymphocyte subset balance in the spleen and thymus and fostered neurodevelopment and neuroimmune balance in the brain. Furthermore, metabolic profiling showed a strong correlation between the metabolites in the serum and colon, supporting the view that B. animalis subsp. lactis pretreatment influences the systemic immune response by modifying the composition and metabolism of the gut microbiota. Overall, the results imply that B. animalis subsp. lactis pretreatment, through the coordinated regulation of colonic and serum metabolites, influences the systemic immune responses of neonatal rats against Salmonella infection.
Collapse
Affiliation(s)
- Chunxiu Lin
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, P. R. China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, P. R. China
| | - Yugui Lin
- Microbiology Laboratory, Zhongshan Bo'ai Hospital, Southern Medical University, Zhongshan 528400, P. R. China
| | - Shunhe Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, P. R. China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, P. R. China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, P. R. China
| | - Jialiang Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, P. R. China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, P. R. China
| | - Xuhua Mao
- Department of Clinical Laboratory, Yixing People's Hospital, Wuxi 214200, P. R. China
| | - Yonghua Zhou
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, P. R. China
| | - Hao Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, P. R. China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, P. R. China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, P. R. China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, P. R. China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, P. R. China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, P. R. China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, P. R. China
| | - Gang Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, P. R. China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, P. R. China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, P. R. China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, P. R. China
| |
Collapse
|
24
|
Hojsak I, Kolaček S. Role of Probiotics in the Treatment and Prevention of Common Gastrointestinal Conditions in Children. Pediatr Gastroenterol Hepatol Nutr 2024; 27:1-14. [PMID: 38249642 PMCID: PMC10796258 DOI: 10.5223/pghn.2024.27.1.1] [Citation(s) in RCA: 1] [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: 04/11/2023] [Revised: 07/10/2023] [Accepted: 08/30/2023] [Indexed: 01/23/2024] Open
Abstract
Probiotics are live microorganisms that confer health benefits to the host when administered in adequate amounts. Although recommendations for probiotic use should be strain-specific, many systematic reviews, including recommendations from different societies, recommend probiotic use in general, providing no relevant information for healthcare professionals regarding which probiotic to recommend for which clinical indication, at what dose, and for how long. This narrative review aimed to present the available evidence on the use of probiotics in the prevention and treatment of common gastrointestinal diseases in children, considering the strain and dose used. Furthermore, this study summarizes the evidence on the possible side effects and quality of products containing probiotics.
Collapse
Affiliation(s)
- Iva Hojsak
- Department of Pediatrics, Referral Center for Pediatric Gastroenterology and Nutrition, Children’s Hospital, Zagreb, Croatia
- Department of Pediatrics, University of Zagreb School of Medicine, Zagreb, Croatia
- Department of Pediatrics, University J.J. Strossmayer School of Medicine, Osijek, Croatia
| | - Sanja Kolaček
- Department of Pediatrics, Referral Center for Pediatric Gastroenterology and Nutrition, Children’s Hospital, Zagreb, Croatia
- Department of Pediatrics, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
25
|
Indrio F, Dargenio VN. Preventing and Treating Colic: An Update. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1449:59-78. [PMID: 39060731 DOI: 10.1007/978-3-031-58572-2_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
Infantile colic (IC) is c is a self-limiting functional gastrointestinal disorder (FGID) with a favorable natural history. Worldwide, IC has a significant impact on many newborns and their families. Although not an indication of an illness, its symptoms are wide and generic and may indicate a potentially serious underlying issue in a tiny percentage of newborns who may require a medical evaluation. The pathogenesis appears to be multifactorial implying a complex relationship between the infant and the environment. One of the most studied theories attributes a key role to the gut microbiota in the pathogenesis of IC. A variety of approaches have been suggested for the clinical management of IC, and several randomized controlled trials have been reported in the literature. Probiotics can change the host's microbiota and positively impact health. They may be able to restore balance and create a better intestinal microbiota landscape since there is mounting evidence that the gut microbial environment of colicky newborns differs from that of healthy infants. In this review, we revise the most commonly studied probiotics and mixtures to treat and prevent IC and the most recent recommendations.
Collapse
Affiliation(s)
- Flavia Indrio
- Department of Experimental Medicine, Università del Salento, Lecce, Italy
| | | |
Collapse
|
26
|
Hojsak I. Probiotics in Functional Gastrointestinal Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1449:157-174. [PMID: 39060737 DOI: 10.1007/978-3-031-58572-2_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
The most frequent functional gastrointestinal disorders (FGID) in children include infantile colic, constipation, functional abdominal pain (FAP), and irritable bowel syndrome (IBS). Unfortunately, treatment options for FGID in children are limited, therefore many dietary interventions have been evaluated, including probiotics. This chapter summarizes currently available evidence and recommendations for probiotic use in the treatment of frequent FGIDs in children. The strongest evidence exists for the use of Limosilactobacillus (L.) reuteri DSM 17938 and Bifidobacterium animalis subsp. lactis BB-12 for the treatment of infantile colic in breastfed infants. Limited but yet encouraging evidence exists for Lacticaseibacillus rhamnosus GG (LGG) for the treatment of IBS and L. reuteri DSM 17938 for FAP.
Collapse
Affiliation(s)
- Iva Hojsak
- Referral Centre for Pediatric Gastroenterology and Nutrition, Children's Hospital Zagreb, Zagreb, Croatia.
- University of Zagreb, School of Medicine, Zagreb, Croatia.
- University J.J. Strossmayer, School of Medicine Osijek, Osijek, Croatia.
| |
Collapse
|
27
|
Gil-Hernández E, Ruiz-González C, Rodriguez-Arrastia M, Ropero-Padilla C, Rueda-Ruzafa L, Sánchez-Labraca N, Roman P. Effect of gut microbiota modulation on sleep: a systematic review and meta-analysis of clinical trials. Nutr Rev 2023; 81:1556-1570. [PMID: 37023468 DOI: 10.1093/nutrit/nuad027] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023] Open
Abstract
CONTEXT A bidirectional relationship between gut microbiota (GM) and circadian rhythms has been proposed. OBJECTIVE The aim of this study was to analyze the efficacy of probiotic or prebiotic intervention on sleep quality and quantity. DATA SOURCES A systematic review and meta-analysis were conducted using the databases PubMed (MEDLINE), Embase, CINAHL, and Web of Science. Only randomized clinical trials written in English or Spanish were considered. DATA EXTRACTION The initial search resulted in 219 articles. Following the removal of duplicates and consideration of the selection criteria, 25 articles were selected for the systematic review and 18 articles for the meta-analysis. DATA ANALYSIS Microbiota modulation was not demonstrated to be associated with significant improvement in sleep quality in the present meta-analysis (P = 0.31). In terms of sleep duration, the meta-analysis found no improvement due to GM modulation (P = 0.43). CONCLUSION The results of this meta-analysis indicate that there is still insufficient evidence to support the relationship between GM modulation and improved sleep quality. While several studies assume that including probiotics in the diet will undoubtedly improve sleep quality, more research is needed to fully understand this phenomenon. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CRD42021245118.
Collapse
Affiliation(s)
| | | | - Miguel Rodriguez-Arrastia
- Faculty of Health Sciences, Department of Nursing Science, Physiotherapy and Medicine, University of Almeria, Almeria, Spain
| | - Carmen Ropero-Padilla
- Faculty of Health Sciences, Department of Nursing Science, Physiotherapy and Medicine, University of Almeria, Almeria, Spain
| | - Lola Rueda-Ruzafa
- Faculty of Health Sciences, Department of Nursing Science, Physiotherapy and Medicine, University of Almeria, Almeria, Spain
| | - Nuria Sánchez-Labraca
- Faculty of Health Sciences, Department of Nursing Science, Physiotherapy and Medicine, University of Almeria, Almeria, Spain
| | - Pablo Roman
- Faculty of Health Sciences, Department of Nursing Science, Physiotherapy and Medicine, University of Almeria, Almeria, Spain
- Health Research Center CEINSA, University of Almeria, Almeria, Spain
- Research Group CTS-1114 Health Sciences, University of Almeria, Almeria, Spain
| |
Collapse
|
28
|
Morales-Cortés VI, Domínguez-Soberanes J, Hernández-Lozano LC, Licon CC, Estevez-Rioja A, Peralta-Contreras M. Sensory characterization of functional guava symbiotic petit cheese product. Heliyon 2023; 9:e21747. [PMID: 38034649 PMCID: PMC10681930 DOI: 10.1016/j.heliyon.2023.e21747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 10/26/2023] [Accepted: 10/26/2023] [Indexed: 12/02/2023] Open
Abstract
The consumption of functional dairy products continues to rise due to consumer needs. This study aimed to develop a dairy guava functional symbiotic petit cheese product that included probiotics (Bifidobacterium animalis subsp. lactis BB-12, Chr. Hansen, Denmark) and prebiotics (inulin), which had adequate organoleptic characteristics. Moreover, adequate physicochemical, microbiological, and sensory characteristics during its shelf life were expected. A pasteurized skim milk curd flavored with a guava pulp was stabilized with gelatin to formulate this product. As sweeteners, iso maltol, erythritol, and Luo Han Guo extract from monk fruit (Siraitia Grosvenorii) were added. The prebiotic used was inulin, and the probiotic (Bifidobacterium animalis subsp. lactis BB-12, Chr. Hansen, Denmark). The product was kept refrigerated (4 °C) during the shelf life of 28 days. For the organoleptic analysis (100 consumers), the evaluations performed were: (1) overall liking (OL), (2) CATA (Check all that apply) testing 19 attributes, and (3) purchase intention was evaluated. Results were analyzed with FIZZ Software Biosystèmes. During shelf life, (1) physicochemical, microbiological, and sensory tests were performed. The product was evaluated as "liked much'' (7.16 out of 9); it was described as a creamy (71 %) natural product (73 %) with a fruity odor (57 %). It could be suitable for marketing because 82 % of the consumers would buy it. The product's probiotic character (over 1 × 106) was established through a microbiological count. On day one, the CFU was found to be 4.15 × 108, and after 28 days, 1.98 × 108 CFU of viable Bifidobacterium animalis subsp. lactis BB-12, leading us to establish its probiotic characteristics. The shelf life was estimated at 21 days.
Collapse
Affiliation(s)
- Victor Iván Morales-Cortés
- Universidad Panamericana. Escuela de Dirección de Negocios Alimentarios. Jose María Escrivá de Balaguer 101, Villas Bonaterra, Aguascalientes, 20296, México
| | - Julieta Domínguez-Soberanes
- Universidad Panamericana. Facultad de Ingeniería. Jose María Escrivá de Balaguer 101, Villas Bonaterra, Aguascalientes, 20296, México
| | - Linda Carolina Hernández-Lozano
- Universidad Panamericana. Escuela de Dirección de Negocios Alimentarios. Jose María Escrivá de Balaguer 101, Villas Bonaterra, Aguascalientes, 20296, México
| | - Carmen C. Licon
- Dairy Products Technology Center, California Polytechnic State University, 1 Grand Avenue, Building 18A, San Luis Obispo, CA, 93407, USA
| | - Antonio Estevez-Rioja
- Universidad Panamericana. Escuela de Dirección de Negocios Alimentarios. Jose María Escrivá de Balaguer 101, Villas Bonaterra, Aguascalientes, 20296, México
| | - Mayeli Peralta-Contreras
- Universidad Panamericana. Escuela de Dirección de Negocios Alimentarios. Jose María Escrivá de Balaguer 101, Villas Bonaterra, Aguascalientes, 20296, México
| |
Collapse
|
29
|
Melsaether C, Høtoft D, Wellejus A, Hermes GDA, Damholt A. Seeding the Infant Gut in Early Life-Effects of Maternal and Infant Seeding with Probiotics on Strain Transfer, Microbiota, and Gastrointestinal Symptoms in Healthy Breastfed Infants. Nutrients 2023; 15:4000. [PMID: 37764787 PMCID: PMC10538230 DOI: 10.3390/nu15184000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/08/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
We investigated the effects of two dosing regimens of two multi-strain probiotic products on the gut microbiota of breastfed infants, including the transfer of the dosed strains and clinical outcomes. In forty-seven dyads, infants were either exposed through maternal intake (MS) of Lactobacillus acidophilus LA-5, Bifidobacterium animalis subsp. lactis BB-12, Lacticaseibacillus rhamnosus LGG, and Bifidobacterium longum subsp. infantis Bifin02 from gestational week thirty-three until four weeks after birth (n = 24) or dosed directly (IS) with the same strains except for LA-5 starting within 24 h after birth until day 28 (n = 23). Infant stool samples were collected on day 0, 14, 28, and 42 after birth. Gastrointestinal symptoms were assessed by parents using an electronic diary. Microbiota composition was determined using 16S rRNA sequencing, and strain recovery was analyzed by qPCR. Notably, 100% of the IS infants were colonized with Bifin02 after 14 days as opposed to only 25% of the MS infants. Mean stool frequency was significantly lower in IS infants compared to MS infants and IS infants had softer stools on day 14, 28, and 42. A significantly steeper slope of progression of inconsolable crying and fussing was observed in MS infants compared to IS infants. In conclusion, direct infant seeding induced a faster increase in fecal bifidobacteria abundancy and Bifin02 recovery compared to dosed through the maternal intake.
Collapse
Affiliation(s)
- Cathrine Melsaether
- Chr. Hansen A/S, Boege Alle 10-12, 2970 Hoersholm, Denmark; (A.W.); (G.D.A.H.); (A.D.)
| | - Diana Høtoft
- Department of Gynecology and Obstetrics, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus, Denmark;
| | - Anja Wellejus
- Chr. Hansen A/S, Boege Alle 10-12, 2970 Hoersholm, Denmark; (A.W.); (G.D.A.H.); (A.D.)
| | - Gerben D. A. Hermes
- Chr. Hansen A/S, Boege Alle 10-12, 2970 Hoersholm, Denmark; (A.W.); (G.D.A.H.); (A.D.)
| | - Anders Damholt
- Chr. Hansen A/S, Boege Alle 10-12, 2970 Hoersholm, Denmark; (A.W.); (G.D.A.H.); (A.D.)
| |
Collapse
|
30
|
Xiang Q, Yan X, Shi W, Li H, Zhou K. Early gut microbiota intervention in premature infants: Application perspectives. J Adv Res 2023; 51:59-72. [PMID: 36372205 PMCID: PMC10491976 DOI: 10.1016/j.jare.2022.11.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/30/2022] [Accepted: 11/05/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Preterm birth is the leading cause of death in children under the age of five. One of the major factors contributing to the high risk of diseases and deaths in premature infants is the incomplete development of the intestinal immune system. The gut microbiota has been widely recognized as a critical factor in promoting the development and function of the intestinal immune system after birth. However, the gut microbiota of premature infants is at high risk of dysbiosis, which is highly associated with adverse effects on the development and education of the early life immune system. Early intervention can modulate the colonization and development of gut microbiota and has a long-term influence on the development of the intestinal immune system. AIM OF REVIEW This review aims to summarize the characterization, interconnection, and underlying mechanism of gut microbiota and intestinal innate immunity in premature infants, and to discuss the status, applicability, safety, and prospects of different intervention strategies in premature infants, thus providing an overview and outlook of the current applications and remaining gaps of early intervention strategies in premature infants. KEY SCIENTIFIC CONCEPTS OF REVIEW This review is focused on three key concepts. Firstly, the gut microbiota of premature infants is at high risk of dysbiosis, resulting in dysfunctional intestinal immune system processes. Secondly, contributing roles of early intervention have been observed in improving the intestinal environment and promoting gut microbiota colonization, which is significant in the development and function of gut immunity in premature infants. Thirdly, different strategies of early intervention, such as probiotics, fecal microbiota transplantation, and nutrients, show different safety, applicability, and outcome in premature infants, and the underlying mechanism is complex and poorly understood.
Collapse
Affiliation(s)
- Quanhang Xiang
- Shenzhen Institute of Respiratory Diseases, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Xudong Yan
- Department of Neonatal Intensive Care Unit, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Wei Shi
- Department of Obstetrics and Gynecology, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Huiping Li
- Department of Respiratory and Critical Care Medicine, the first affiliated hospital of Southern University of Science and Technology of China, Shenzhen People's Hospital, Shenzhen, China; The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China
| | - Kai Zhou
- Shenzhen Institute of Respiratory Diseases, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China; The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
31
|
Cudris-Torres L, Alpi SV, Barrios-Núñez Á, Gaviria Arrieta N, Mejía Gutiérrez J, Alvis Barranco L, Rios-Carlys G, Cuenca-Calderón SE, Bermúdez V, Hernández-Lalinde J, Riveira Zuleta CA, Bahamón MJ, Álvarez Herrera JS. Quality of life in the older adults: The protective role of self-efficacy in adequate coping in patients with chronic diseases. Front Psychol 2023; 14:1106563. [PMID: 37089743 PMCID: PMC10117781 DOI: 10.3389/fpsyg.2023.1106563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 03/13/2023] [Indexed: 04/09/2023] Open
Abstract
The purpose of the present study was to establish the association between self-efficacy, perception of disease, emotional regulation, and fatigue and the health-related quality of life in older adults living in the departments of Cesar and Atlántico in Colombia and who have been diagnosed with a chronic disease. The participants were 325 older adults of both sexes, with literacy and no presence of cognitive impairment in the Mini-Mental State Examination (MMSE); A non-probabilistic sampling was carried out. We used the MOS-SF-36 questionnaire, the Brief Illness Perception Questionnaire scale for measuring the perception of disease, the Stanford Patient Education Research Center’s Chronic Disease Self self-efficacy questionnaire for chronic patients, the Difficulties in Emotional Regulation Scale, and the Fatigue Severity Questionnaire as measurement instruments. The design was non-experimental cross-sectional with a correlational scope. The results indicate that self-efficacy, disease perception, emotional regulation and severity of fatigue are variables that could impact the physical function of quality of life, confirming that self-efficacy would work as a factor that decreases the probability that a participant score low on this dimension of quality of life. On the other hand, both the perception of the disease and the severity of fatigue were identified as factors that probably negatively influence quality of life.
Collapse
Affiliation(s)
- Lorena Cudris-Torres
- Programa de Psicología, Fundación Universitaria del Área Andina, Valledupar, Colombia
- *Correspondence: Lorena Cudris-Torres,
| | | | | | | | | | | | - Gerson Rios-Carlys
- Programa de Psicología, Universidad Popular del Cesar, Valledupar, Colombia
| | | | - Valmore Bermúdez
- Centro de Investigaciones en Ciencias de la Vida, Universidad Simón Bolívar, Barranquilla, Colombia
| | | | | | | | | |
Collapse
|
32
|
Probiotics for the Management of Pediatric Gastrointestinal Disorders: Position Paper of the ESPGHAN Special Interest Group on Gut Microbiota and Modifications. J Pediatr Gastroenterol Nutr 2023; 76:232-247. [PMID: 36219218 DOI: 10.1097/mpg.0000000000003633] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Probiotics, defined as live microorganisms that, when administered in adequate amounts, confer a health benefit on the host, are widely used despite uncertainty regarding their efficacy and discordant recommendations about their use. The European Society for Paediatric Gastroenterology, Hepatology and Nutrition (ESPGHAN) Special Interest Group on Gut Microbiota and Modifications provides updated recommendations for the use of probiotics for the management of selected pediatric gastrointestinal disorders. METHODS All systematic reviews and/or meta-analyses, as well as subsequently published randomized controlled trials (RCTs) (until December 2021), that compared the use of probiotics in all delivery vehicles and formulations, at any dose, with no probiotic (ie, placebo or no treatment), were eligible for inclusion. The recommendations were formulated only if at least 2 RCTs on a similar well-defined probiotic strain were available. The modified Delphi process was used to establish consensus on the recommendations. RESULTS Recommendations for the use of specific probiotic strains were made for the management of acute gastroenteritis, prevention of antibiotic-associated diarrhea, nosocomial diarrhea and necrotizing enterocolitis, management of Helicobacter pylori infection, and management of functional abdominal pain disorders and infant colic. CONCLUSIONS Despite evidence to support the use of specific probiotics in some clinical situations, further studies confirming the effect(s) and defining the type, dose, and timing of probiotics are still often required. The use of probiotics with no documented health benefits should be discouraged.
Collapse
|
33
|
Liu F, Smith AD, Wang TTY, Pham Q, Cheung L, Yang H, Li RW. Biological pathways via which the anthocyanin malvidin alleviated the murine colitis induced by Citrobacter rodentium. Food Funct 2023; 14:1048-1061. [PMID: 36562464 DOI: 10.1039/d2fo02873e] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Enteropathogenic E. coli (EPEC) is a causal agent for diarrheal diseases and contributes to morbidity and mortality in children under the age of five years. The emergence and rapid spread of antibiotic resistant EPEC strains necessitate the search for novel alternatives to antibiotics. In this study, we used Citrobacter rodentium, a natural mouse pathogen that mimics many aspects of human EPEC infections, to investigate the antimicrobial properties of the blueberry anthocyanin malvidin 3-glucoside (MG) using a multi-omics approach. MG supplementation reversed the bodyweight loss induced by C. rodentium infection and improved colonic hyperplasia and histopathological scores. In the colon tissue, MG supplementation significantly increased the expression of Hace1, a key regulator of TNFα-driven signaling, and impacted multiple pathways, such as TGFβ signaling. MG partially restored C. rodentium-induced microbial dysbiosis and significantly enhanced the abundance of the probiotic Bifidobacterium animalis. Moreover, MG disrupted the interactions of E. coli with other gut microbes. MG significantly mediated several host- and microbiota-derived metabolites, such as cytosine, ureidopropionic acid, and glutaric acid. MG normalized the bioactive lipid oleoylethanolamine, a member of the endocannabinoid system, from the dysregulated level in infected mice, directly contributing to its overall beneficial effects. Our findings provided novel insights into molecular processes via which the flavonoid malvidin exerts its biological effects in the gastrointestinal tract.
Collapse
Affiliation(s)
- Fang Liu
- College of Public Health, Zhengzhou University, Zhengzhou, China.
| | - Allen D Smith
- USDA-ARS, Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Beltsville, MD, USA
| | - Thomas T Y Wang
- USDA-ARS, Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Beltsville, MD, USA
| | - Quynhchi Pham
- USDA-ARS, Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Beltsville, MD, USA
| | - Lumei Cheung
- USDA-ARS, Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Beltsville, MD, USA
| | - Haiyan Yang
- College of Public Health, Zhengzhou University, Zhengzhou, China.
| | - Robert W Li
- USDA-ARS, Animal Parasitic Diseases Laboratory, Beltsville, MD, USA.
| |
Collapse
|
34
|
Sankova MV, Nikolenko VN, Sankov SV, Sinelnikov MY. SARS-CoV-2 and microbiome. AUTOIMMUNITY, COVID-19, POST-COVID19 SYNDROME AND COVID-19 VACCINATION 2023:279-337. [DOI: 10.1016/b978-0-443-18566-3.00023-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
35
|
Tyszka M, Maciejewska-Markiewicz D, Biliński J, Lubas A, Stachowska E, Basak GW. Increased Intestinal Permeability and Stool Zonulin, Calprotectin and Beta-Defensin-2 Concentrations in Allogenic Hematopoietic Cell Transplantation Recipients. Int J Mol Sci 2022; 23:ijms232415962. [PMID: 36555600 PMCID: PMC9781277 DOI: 10.3390/ijms232415962] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/08/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Significant progress has been made in understanding the connection between intestinal barrier function and allogenic hematopoietic cell transplantation (allo-HCT) recipients' outcomes. The purpose of this study was to further evaluate gut barrier permeability and other potential intestinal barrier disruption markers in the allo-HCT setting. Fifty-one patients were enrolled in the study. Intestinal permeability was assessed with the sugar absorption test and faecal concentrations of the zonulin, calprotectin and beta-defensin-2 levels in the peri-transplantation period. Most patients undergoing allo-HCT in our department had a disrupted intestinal barrier at the baseline, which was associated with older age and higher Hematopoietic Cell Transplantation-specific Comorbidity Index (HCT-CI). Regardless of this, we observed a further increase in gut barrier permeability after allo-HCT in most patients. However, there was no association between permeability assay and other markers (zonulin, calprotectin and beta-defensin-2). Patients with acute GVHD had significantly higher median calprotectin concentrations after allo-HCT compared with the patients without this complication. Our findings indicate that gut barrier damage develops prior to allo-HCT with progression after the procedure and precedes further complications, but did not prove other markers to be useful surrogates of intestinal permeability.
Collapse
Affiliation(s)
- Martyna Tyszka
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
- Correspondence: (M.T.); (D.M.-M.)
| | - Dominika Maciejewska-Markiewicz
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland
- Correspondence: (M.T.); (D.M.-M.)
| | - Jarosław Biliński
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Arkadiusz Lubas
- Department of Internal Medicine, Nephrology and Dialysis, Military Institute of Medicine, 04-141 Warsaw, Poland
| | - Ewa Stachowska
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland
| | - Grzegorz W. Basak
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
| |
Collapse
|
36
|
Sheldon JM, Alonso N. The Therapeutic Benefits of Single and Multi-Strain Probiotics on Mean Daily Crying Time and Key Inflammatory Markers in Infantile Colic. Cureus 2022; 14:e28363. [PMID: 36168359 PMCID: PMC9506670 DOI: 10.7759/cureus.28363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Infantile colic is a functional gastrointestinal disorder in which a healthy infant displays paroxysms of intense crying or fussiness. Although this condition is self-limited, it causes significant distress for parents and may be linked to long-term health concerns for children. The microbiome of infants with colic has been correlated with increased dysbiosis or imbalance of commensal bacteria. This dysbiosis may ultimately lead to changes in infants’ immunological profiles, favoring markers linked to inflammation, including specific cytokines, calprotectin, and genetic markers. Therapeutic regimens such as probiotics may be helpful in modifying the gut microbial composition, thereby influencing the presence of inflammatory markers and potentially reducing colic symptoms in infants. This review provides a summary of the findings from 10 randomized, placebo-controlled, double-blinded studies conducted in the past five years with the aim of examining the potential therapeutic benefits of probiotics in infantile colic. The articles were selected through PubMed and Google Scholar using the keywords infantile colic, microbiome, probiotics, cytokines, dysbiosis, inflammatory markers, and lactobacilli. We summarize the results of these studies to explore the potential anti-inflammatory therapeutic benefits of single and multi-strain probiotic formulations on daily crying time and key inflammatory markers in infants with colic. The research largely shows the beneficial role of probiotics, largely of the lactobacillus genus, in the reduction of colic symptoms and the reduction of key inflammatory markers. However, some studies demonstrated an insignificant effect of certain probiotic strains in symptom management. Further research is necessary to better understand the anti-inflammatory properties of probiotics and determine the role this could have on the manifestation of colic in infants.
Collapse
|
37
|
Łoniewski I, Skonieczna-Żydecka K, Stachowska L, Fraszczyk-Tousty M, Tousty P, Łoniewska B. Breastfeeding Affects Concentration of Faecal Short Chain Fatty Acids During the First Year of Life: Results of the Systematic Review and Meta-Analysis. Front Nutr 2022; 9:939194. [PMID: 35898706 PMCID: PMC9310010 DOI: 10.3389/fnut.2022.939194] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 06/21/2022] [Indexed: 11/21/2022] Open
Abstract
Short chain fatty acids (SCFAs) are important metabolites of the gut microbiota. It has been shown that the microbiota and its metabolic activity in children are highly influenced by the type of diet and age. Our aim was to analyse the concentration of fecal SCFAs over two years of life and to evaluate the influence of feeding method on the content of these compounds in feces. We searched PubMed/MEDLINE/Embase/Ebsco/Cinahl/Web of Science from the database inception to 02/23/2021 without language restriction for observational studies that included an analysis of the concentration of fecal SCFAs in healthy children up to 3 years of age. The primary outcome measures-mean concentrations-were calculated. We performed a random-effects meta-analysis of outcomes for which ≥2 studies provided data. A subgroup analysis was related to the type of feeding (breast milk vs. formula vs. mixed feeding) and the time of analysis (time after birth). The initial search yielded 536 hits. We reviewed 79 full-text articles and finally included 41 studies (n = 2,457 SCFA analyses) in the meta-analysis. We found that concentrations of propionate and butyrate differed significantly in breastfed infants with respect to time after birth. In infants artificially fed up to 1 month of age, the concentration of propionic acid, butyric acid, and all other SCFAs is higher, and acetic acid is lower. At 1–3 months of age, a higher concentration of only propionic acid was observed. At the age of 3–6 months, artificial feeding leads to a higher concentration of butyric acid and the sum of SCFAs. We concluded that the type of feeding influences the content of SCFAs in feces in the first months of life. However, there is a need for long-term evaluation of the impact of the observed differences on health later in life and for standardization of analytical methods and procedures for the study of SCFAs in young children. These data will be of great help to other researchers in analyzing the relationships between fecal SCFAs and various physiologic and pathologic conditions in early life and possibly their impact on health in adulthood.
Collapse
Affiliation(s)
- Igor Łoniewski
- Department of Biochemical Science, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Karolina Skonieczna-Żydecka
- Department of Biochemical Science, Pomeranian Medical University in Szczecin, Szczecin, Poland
- *Correspondence: Karolina Skonieczna-Żydecka
| | - Laura Stachowska
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | | | - Piotr Tousty
- Department of Obstetrics and Gynecology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Beata Łoniewska
- Department of Neonatal Diseases, Pomeranian Medical University in Szczecin, Szczecin, Poland
| |
Collapse
|
38
|
Johnson JM, Adams ED. The Gastrointestinal Microbiome in Infant Colic: A Scoping Review. MCN Am J Matern Child Nurs 2022; 47:195-206. [PMID: 35352686 DOI: 10.1097/nmc.0000000000000832] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
ABSTRACT The significant crying of infantile colic adds stress to the infant and their family, yet it has no recognized etiology. Gastrointestinal health problems and dysfunction have been suspected in the etiology of colic. Disruptions to the microbiome colonization of the gastrointestinal system may lead to excess gas and inflammation that are associated with the crying of colic. Infants with colic have increased colonization with gas-producing bacteria, like Escherichia coli and Klebsiella , and they have lower colonization of anti-inflammatory bacteria, like Bifidobacterium and Lactobacillus . Colic is known to self-resolve around 3 months of age. However, few researchers have investigated how the microbiome may be changing at colic's natural resolution without the intervention of a probiotic. With a better understanding of what leads to colic's self-resolution, future researchers may be able to identify more effective therapies for colic prevention or treatment. This scoping review presents the collective evidence from 21 original, primary research articles on what is known about the gastrointestinal microbiome at colic onset and resolution.
Collapse
|
39
|
Han X, Guo J, Qin Y, Huang W, You Y, Zhan J. Dietary regulation of the SIgA-gut microbiota interaction. Crit Rev Food Sci Nutr 2022; 63:6379-6392. [PMID: 35125055 DOI: 10.1080/10408398.2022.2031097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Gut microbiota (GM) is essential for host health, and changes in the GM are related to the development of various diseases. Recently, secretory immunoglobulin A (SIgA), the most abundant immunoglobulin isotype in the intestinal mucosa, has been found to play an essential role in controlling GM. SIgA dysfunction can lead to changes in the GM and is associated with the development of various GM-related diseases. Although in early stage, recent studies have shown that assorted dietary interventions, including vitamins, amino acids, fatty acids, polyphenols, oligo/polysaccharides, and probiotics, can influence the intestinal SIgA response and SIgA-GM interaction. Dietary intervention can enhance the SIgA response by directly regulating it (from top to bottom) or by regulating the GM structure or gene expression (from bottom to top). Furthermore, intensive studies involving the particular influence of dietary intervention on SIgA-binding to the GM and SIgA repertoire and the precise regulation of the SIgA response via dietary intervention are still exceedingly scarce and merit further consideration. This review summarizes the existing knowledge and (possible) mechanisms of the influence of dietary intervention on the SIgA-GM interaction. Key issues are considered, and the approaches in addressing these issues in future studies are also discussed.
Collapse
Affiliation(s)
- Xue Han
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing
| | - Jielong Guo
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Yue Qin
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Weidong Huang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Yilin You
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Jicheng Zhan
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| |
Collapse
|
40
|
Mao K, Gao J, Wang X, Li X, Geng S, Zhang T, Sadiq FA, Sang Y. Bifidobacterium animalis subsp. lactis BB-12 Has Effect Against Obesity by Regulating Gut Microbiota in Two Phases in Human Microbiota-Associated Rats. Front Nutr 2022; 8:811619. [PMID: 35083265 PMCID: PMC8784422 DOI: 10.3389/fnut.2021.811619] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/08/2021] [Indexed: 12/21/2022] Open
Abstract
Bifidobacterium animalis subsp. lactis BB-12 (BB-12) is an extensively studied probiotics species, which has been reported to improve the human gut microbiota. This study aimed to confirm the effects of BB-12 on high-fat diet (HFD)-induced gut microbiota disorders. The probiotic BB-12 was consumed by human microbiota-associated rats and changes in gut microbiota were compared using next generation sequencing of the fecal samples collected from the normal chow group, the HFD group, and the BB-12-supplemented group. The enterotypes switched from Prevotella dominant to Akkermansia dominant as a result of switching diet from normal chow to HFD. BB-12 conferred protection on the gut microbiota composition of the rats by increasing the abundance of Prevotella and decreasing the abundance of Clostridium, Blautia, and Bacteroides in 0-3 weeks. In addition, Prevotella-dominant enterotype was maintained, which provides improve obesity effects. A decrease in body weight and the Firmicutes/Bacteroidetes ratio were also observed at week 3. While in 4-8 weeks, the enrichment of short-chain fatty acids-producing bacteria such as Eubacterium and Parabacteroides and probiotics such as Bifidobacterium was observed. The results revealed that BB-12 against obesity by regulating gut microbiota in two phases. After a short-term intervention, BB-12 supplementation suppressed the transition from the healthy to obesity state by protecting Prevotella-dominant enterotype, whereas after a long-term intervention, BB-12 ameliorates obesity by enriching beneficial bacteria in the gut.
Collapse
Affiliation(s)
- Kemin Mao
- Department of Food Science and Technology, Hebei Agricultural University, Baoding, China
| | - Jie Gao
- Department of Food Science and Technology, Hebei Agricultural University, Baoding, China
| | - Xianghong Wang
- Department of Food Science and Technology, Hebei Agricultural University, Baoding, China
| | - Xiyu Li
- Department of Food Science and Technology, Hebei Agricultural University, Baoding, China
| | - Shuo Geng
- Department of Food Science and Technology, Hebei Agricultural University, Baoding, China
| | - Tuo Zhang
- Department of Food Science and Technology, Hebei Agricultural University, Baoding, China
| | | | - Yaxin Sang
- Department of Food Science and Technology, Hebei Agricultural University, Baoding, China
| |
Collapse
|
41
|
Astó E, Huedo P, Altadill T, Aguiló García M, Sticco M, Perez M, Espadaler-Mazo J. Probiotic Properties of Bifidobacterium longum KABP042 and Pediococcus pentosaceus KABP041 Show Potential to Counteract Functional Gastrointestinal Disorders in an Observational Pilot Trial in Infants. Front Microbiol 2022; 12:741391. [PMID: 35095783 PMCID: PMC8790238 DOI: 10.3389/fmicb.2021.741391] [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: 07/14/2021] [Accepted: 12/23/2021] [Indexed: 12/22/2022] Open
Abstract
Functional gastrointestinal disorders (FGIDs) are a common concern during the first year of life. Recognized as gut-brain axis disorders by Rome IV criteria, FGIDs etiology is linked to altered gut-brain interaction, intestinal physiology, and microbiota. In this regard, probiotics have emerged as a promising therapy for infant FGIDs. In this study, we have investigated the probiotic potential of the strains Bifidobacterium longum KABP042 and Pediococcus pentosaceus KABP041-isolated from healthy children's feces-in the treatment of FGIDs. To this scope, genome sequences of both strains were obtained and subjected to in silico analyses. No virulence factors were detected for any strain and only the non-transferable erm(49) gene, which confers resistance to erythromycin and clindamycin, was identified in the genome of B. longum KABP042. Safety of both strains was confirmed by acute oral toxicity in rats. In vitro characterization revealed that the strains tolerate gastric and bile challenges and display a great adhesion capacity to human intestinal cells. The two strains mediate adhesion by different mechanisms and, when combined, synergically induce the expression of Caco-2 tight junction proteins. Moreover, growth inhibition experiments demonstrated the ability of the two strains alone and in combination to antagonize diverse Gram-negative and Gram-positive bacterial pathogens during sessile and planktonic growth. Pathogens' inhibition was mostly mediated by the production of organic acids, but neutralization experiments strongly suggested the presence of additional antimicrobial compounds in probiotic culture supernatants such as the bacteriocin Lantibiotic B, whose gene was detected in the genome of B. longum KABP042. Finally, an exploratory, observational, pilot study involving 36 infants diagnosed with at least one FGID (infant colic and/or functional constipation) showed the probiotic formula was well tolerated and FGID severity was significantly reduced after 14 days of treatment with the 2 strains. Overall, this work provides evidence of the probiotic and synergic properties of strains B. longum KABP042 and P. pentosaceus KABP041, and of their potential to treat pediatric FGIDs. Clinical Trial Registration: [www.ClinicalTrials.gov], [identifier NCT04944628].
Collapse
Affiliation(s)
- Erola Astó
- R&D Department, AB-Biotics S.A. (Part of Kaneka Corporation), Barcelona, Spain
- Basic Sciences Department, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Pol Huedo
- R&D Department, AB-Biotics S.A. (Part of Kaneka Corporation), Barcelona, Spain
| | - Tatiana Altadill
- R&D Department, AB-Biotics S.A. (Part of Kaneka Corporation), Barcelona, Spain
- Basic Sciences Department, Universitat Internacional de Catalunya, Barcelona, Spain
| | | | - Maura Sticco
- Pediatric Primary Care Local Health Authority, ASL Caserta, Caserta, Italy
| | - Marta Perez
- R&D Department, AB-Biotics S.A. (Part of Kaneka Corporation), Barcelona, Spain
| | | |
Collapse
|
42
|
Shehata HR, Kiefer A, Morovic W, Newmaster SG. Locked Nucleic Acid Hydrolysis Probes for the Specific Identification of Probiotic Strains Bifidobacterium animalis subsp. lactis DSM 15954 and Bi-07™. Front Microbiol 2022; 12:801795. [PMID: 35003031 PMCID: PMC8733699 DOI: 10.3389/fmicb.2021.801795] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/25/2021] [Indexed: 11/13/2022] Open
Abstract
Probiotic health benefits are now well-recognized to be strain specific. Probiotic strain characterization and identification is thus important in clinical research and in the probiotic industry. This is becoming especially important with reports of probiotic products failing to meet the declared strain content, potentially compromising their efficacy. Availability of reliable identification methods is essential for strain authentication during discovery, evaluation and commercialization of a probiotic strain. This study aims to develop identification methods for strains Bifidobacterium animalis subsp. lactis DSM 15954 and Bi-07 (Bi-07™) based on real-time PCR, targeting single nucleotide polymorphisms (SNPs). The SNPs were targeted by PCR assays with locked nucleic acid (LNA) probes, which is a novel application in probiotic identification. The assays were then validated following the guidelines for validating qualitative real-time PCR assays. Each assay was evaluated for specificity against 22 non-target strains including closely related Bifidobacterium animalis subsp. lactis strains and were found to achieve 100% true positive and 0% false positive rates. To determine reaction sensitivity and efficiency, three standard curves were established for each strain. Reaction efficiency values were 86, 91, and 90% (R square values > 0.99), and 87, 84, and 86% (R square values > 0.98) for B. animalis subsp. lactis DSM 15954 and Bi-07 assays, respectively. The limit of detection (LOD) was 5.0 picograms and 0.5 picograms of DNA for DSM 15954 and Bi-07 assays, respectively. Each assay was evaluated for accuracy using five samples tested at three different DNA concentrations and both assays proved to be highly repeatable and reproducible. Standard deviation of Cq values between two replicates was always below 1.38 and below 1.68 for DSM 15954 and Bi-07 assays, respectively. The assays proved to be applicable to mono-strain and multi-strain samples as well as for samples in various matrices of foods or dietary supplement ingredients. Overall, the methods demonstrated high specificity, sensitivity, efficiency and precision and broad applicability to sample, matrix and machine types. These methods facilitate strain level identification of the highly monophyletic strains B. animalis subsp. lactis DSM 15954 and Bi-07 to ensure probiotic efficacy and provide a strategy to identify other closely related probiotics organisms.
Collapse
Affiliation(s)
- Hanan R Shehata
- Natural Health Product Research Alliance, College of Biological Science, University of Guelph, Guelph, ON, Canada.,Department of Microbiology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Anthony Kiefer
- IFF Health & Biosciences, International Flavors and Fragrances, Inc., Madison, WI, United States
| | - Wesley Morovic
- IFF Health & Biosciences, International Flavors and Fragrances, Inc., Madison, WI, United States
| | - Steven G Newmaster
- Natural Health Product Research Alliance, College of Biological Science, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
43
|
Probiotics in the treatment of infantile colic: a meta-analysis of randomized controlled trials. NUTR HOSP 2022; 39:1135-1143. [DOI: 10.20960/nh.04011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
|
44
|
Chen K, Zhang G, Xie H, You L, Li H, Zhang Y, Du C, Xu S, Melsaether C, Yuan S. Efficacy of Bifidobacterium animalis subsp. lactis, BB-12 ® on infant colic - a randomised, double-blinded, placebo-controlled study. Benef Microbes 2021; 12:531-540. [PMID: 34550055 DOI: 10.3920/bm2020.0233] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
To evaluate the administration of Bifidobacterium animalis subsp. lactis, BB-12® (BB-12) on infant colic in breastfed infants, a double-blind, placebo-controlled randomised study was conducted in Chengdu, China from April 2016 to October 2017 with 192 full-term infants less than 3 months of age and meeting the ROME III criteria for infant colic. After a 1-week run-in the infants were randomly assigned to receive daily BB-12 (1×109 cfu/day) or placebo for 3 weeks. Crying/fussing time were recorded using a 24 h structured diary. The primary endpoint was the proportion of infants achieving a reduction in crying and fussing time of ≥50% from baseline. Parent's/caregiver's health related quality of life was measured using a modified PedsQL™ 2.0 Family Impact Module and immunological biomarkers were evaluated from faecal samples at baseline and after the 21-day intervention. The percentage of infants achieving a reduction in the daily crying/fussing time ≥50% after the 21-day intervention was significantly higher in the infants supplemented with BB-12 (P<0.001). The mean number of crying episodes was significantly reduced in the BB-12 group compared to the placebo group (10.0±3.0 to 5.0±1.87 vs 10.5±2.6 to 7.5±2.8, respectively) (P<0.001) and the mean daily sleep duration was markedly increased from baseline to end of intervention in the BB-12 group compared to the infants in the placebo group (60.7±104.0 vs 31.9±102.7 min/day, respectively) (P<0.001). The faecal levels of human beta defensin 2, cathelicidin, slgA, calprotectin and butyrate were statistically higher in the BB-12 group compared to the placebo group after the 21-day intervention. At the end of the intervention the parent's/caregiver's physical, emotional and social functioning scores were significantly higher for the BB-12 group compared to the placebo group (all P<0.05). Supplementation of BB-12 is effective in reducing crying and fussing in infants diagnosed with infant colic.
Collapse
Affiliation(s)
- K Chen
- Department of Nutrition, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 1617, Riyue Avenue, Qingyang District, Chengdu, 6100131, China P.R.,Department of Child Health Care, Chengdu New Century Women's and Children's Hospital, No.77, Baojia Lane, Qingyang District, Chengdu, China P.R
| | - G Zhang
- Department of Pediatric Intensive Care Unit, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 1617, Riyue Avenue, Qingyang District, Chengdu, China P.R
| | - H Xie
- Department of Pediatrics, Dayi Maternal and Child Health Care Hospital, No. 539, Inner Mongolia Avenue, Jinyuan Town, Dayi County, Chengdu, China P.R
| | - L You
- Department of Child Health Care, Nanxin Community Health Service Center, N0. 168, Guanghe 1st Street, Wuhou District, Chengdu, China P.R
| | - H Li
- Department of Child Health Care, Qingbaijiang Maternal and Child Health Care Hospital, No.87, Qingjiang South Road, Qingbaijiang District, Chengdu, China P.R
| | - Y Zhang
- Department of Child Health Care, Jinniu Maternal and Child Health Care Hospital, No.12, Changyue Road, Jinniu District, Chengdu, China P.R
| | - C Du
- Department of Child Health Care, Longquanyi Maternal and Child Health Care Hospital, No.383, Yuyang Road, Longquanyi District, Chengdu, China P.R
| | - S Xu
- Department of Child Health Care, Huili Maternal and Child Health Care Hospital, No. 41, Jindai Road West Section, Guoyuan Township, Huili County, Xichang, China P.R
| | - C Melsaether
- Chr. Hansen A/S, HH Clinical Development, Kogle Alle 6, 2970 Hoersholm, Denmark
| | - S Yuan
- Department of Nutrition, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 1617, Riyue Avenue, Qingyang District, Chengdu, 6100131, China P.R
| |
Collapse
|
45
|
Silva SF, Rocha RS, Esmerino EA, Pimentel TC, Gomes da Cruz A, Rodrigues Anjos CA. Impact of different modified atmosphere packaging on quality parameters and probiotic survival during storage of Minas Frescal cheese. FOOD BIOSCI 2021. [DOI: 10.1016/j.fbio.2021.101338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
46
|
|
47
|
Freedman SB, Horne R, Johnson-Henry K, Xie J, Williamson-Urquhart S, Chui L, Pang XL, Lee B, Schuh S, Finkelstein Y, Gouin S, Farion KJ, Poonai N, Hurley K, Schnadower D, Sherman PM, Pediatric Emergency Research Canada Probiotic Regimen for Outpatient Gastroenteritis Utility of Treatment (PROGUT) Trial Group. Probiotic stool secretory immunoglobulin A modulation in children with gastroenteritis: a randomized clinical trial. Am J Clin Nutr 2021; 113:905-914. [PMID: 34269370 PMCID: PMC8023833 DOI: 10.1093/ajcn/nqaa369] [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: 08/06/2020] [Accepted: 11/13/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND We previously conducted the Probiotic Regimen for Outpatient Gastroenteritis Utility of Treatment (PROGUT) study, which identified no improvements in children with acute gastroenteritis (AGE) administered a probiotic. However, the aforementioned study did not evaluate immunomodulatory benefits. OBJECTIVES The object of this study was to determine if stool secretory immunoglobulin A (sIgA) concentrations in children with AGE increase more among participants administered a Lactobacillus rhamnosus/helveticus probiotic compared with those administered placebo. METHODS This a priori planned multicenter, randomized, double-blinded, placebo-controlled ancillary study enrolled children presenting for emergency care who received a 5-d probiotic or placebo course. Participants submitted stool specimens on days 0, 5, and 28. The primary endpoint was the change in stool sIgA concentrations on day 5 compared with baseline. RESULTS A total of 133 (n = 66 probiotic, 67 placebo) of 886 PROGUT participants (15.0%) provided all 3 specimens. Median stool sIgA concentrations did not differ between the probiotic and placebo groups at any of the study time points: day 0 median (IQR): 1999 (768, 4071) compared with 2198 (702, 5278) (P = 0.27, Cohen's d = 0.17); day 5: 2505 (1111, 5310) compared with 3207 (982, 7080) (P = 0.19, Cohen's d = 0.16); and day 28: 1377 (697, 2248) compared with 1779 (660, 3977) (P = 0.27, Cohen's d = 0.19), respectively. When comparing measured sIgA concentrations between days 0 and 5, we found no treatment allocation effects [β: -0.24 (-0.65, 0.18); P = 0.26] or interaction between treatment and specimen collection day [β: -0.003 (-0.09, 0.09); P = 0.95]. Although stool sIgA decreased between day 5 and day 28 within both groups (P < 0.001), there were no differences between the probiotic and placebo groups in the median changes in sIgA concentrations when comparing day 0 to day 5 median (IQR) [500 (-1135, 2362) compared with 362 (-1122, 4256); P = 0.77, Cohen's d = 0.075] and day 5 to day 28 [-1035 (-3130, 499) compared with -1260 (-4437, 843); P = 0.70, Cohen's d = 0.067], respectively. CONCLUSIONS We found no effect of an L. rhamnosus/helveticus probiotic, relative to placebo, on stool IgA concentrations. This trial was registered at clinicaltrials.gov as NCT01853124.
Collapse
Affiliation(s)
- Stephen B Freedman
- Sections of Pediatric Emergency Medicine and Gastroenterology, Department of Pediatrics, Alberta Children's Hospital, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Department of Emergency Medicine, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Rachael Horne
- Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Kathene Johnson-Henry
- Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Jianling Xie
- Section of Pediatric Emergency Medicine, Department of Pediatrics, Alberta Children's Hospital, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Sarah Williamson-Urquhart
- Section of Pediatric Emergency Medicine, Department of Pediatrics, Alberta Children's Hospital, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Linda Chui
- Alberta Precision Laboratories - ProvLab, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Canada
| | - Xiao-Li Pang
- Alberta Precision Laboratories - ProvLab, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Canada
| | - Bonita Lee
- Department of Pediatrics, Faculty of Medicine & Dentistry, Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada
| | - Suzanne Schuh
- Division of Emergency Medicine, Department of Paediatrics, Hospital for Sick Children, Toronto, Canada
| | - Yaron Finkelstein
- Divisions of Emergency Medicine and Clinical Pharmacology and Toxicology, Research Institute, Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Serge Gouin
- Departments of Pediatric Emergency Medicine & Pediatrics, CHU Sainte-Justine, Université de Montréal, Montreal, Canada
| | - Ken J Farion
- Departments of Pediatrics and Emergency Medicine, and Pediatric Emergency Department, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, Canada
| | - Naveen Poonai
- Division of Pediatric Emergency Medicine, Departments of Pediatrics, Internal Medicine, Epidemiology & Biostatistics, Schulich School of Medicine and Dentistry, London, Canada
| | - Katrina Hurley
- Division of Paediatric Emergency Medicine, Dalhousie University, Halifax, Canada
| | - David Schnadower
- Division of Emergency Medicine, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Philip M Sherman
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Paediatrics, and the Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Canada
| | | |
Collapse
|
48
|
Piątek J, Bernatek M, Krauss H, Wojciechowska M, Chęcińska-Maciejewska Z, Kaczmarek P, Sommermeyer H. Effects of a nine-strain bacterial synbiotic compared to simethicone in colicky babies - an open-label randomised study. Benef Microbes 2021; 12:249-257. [PMID: 33765904 DOI: 10.3920/bm2020.0160] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The aim of the study was to determine effects of administration of simethicone and a multi-strain synbiotic on the crying behaviour of colicky babies. The study design consisted of an open-label, two parallel treatment group study involving 87 infants aged 3-6 weeks with infantile colic (defined as crying episodes lasting 3 or more hours per day and occurring at least 3 days per week within 3 weeks prior to enrolment) randomly, unequally [1:1.5] assigned to receive simethicone (n=33) or a multi-strain synbiotic (n=54) orally for 4 weeks. The multi-strain synbiotic contained Lactobacillus acidophilus LA-14, Lacticaseibacillus casei R0215, Lacticaseibacillus paracasei Lp-115, Lacticaseibacillus rhamnosus GG, Ligilactobacillus salivarius Ls-33, Bifidobacterium lactis Bl-04, Bifidobacterium bifidum R0071, Bifidobacterium longum R0175 and fructooligosaccharides). Primary outcome measures were the responder rates (effect ≥50% reduction from baseline) of the measures 'crying days last 3 weeks', 'average evening crying duration last 3 weeks' and 'reduction of average number of crying phases per day last three weeks' at the end of treatment. The study is registered at ClinicalTrials.gov under NCT04487834. Significantly higher responder rates (effect ≥50% reduction from baseline) of the multi-strain synbiotic compared to simethicone were found for the measures 'crying days last 3 weeks' (72% vs 18%, P<0.0001) and 'average evening crying duration last 3 weeks' (85% vs 39%, P=0.0001). No significant difference was found for the measure 'reduction of average number of crying phases per day last three weeks' (50% vs 42%, P=0.4852). No adverse effects were reported for the two treatment groups. Based on these results, the multi-strain synbiotic can be considered as an interesting therapeutic possibility for the treatment of infantile colic, worthwhile to be investigated further in non-clinical and clinical studies.
Collapse
Affiliation(s)
- J Piątek
- Department of Medicine, The President Stanisław Wojciechowski State University of Applied Sciences in Kalisz, Nowy Šwiat 4, 62-800 Kalisz, Poland
| | - M Bernatek
- Department of Medicine, The President Stanisław Wojciechowski State University of Applied Sciences in Kalisz, Nowy Šwiat 4, 62-800 Kalisz, Poland
| | - H Krauss
- Department of Medicine, The President Stanisław Wojciechowski State University of Applied Sciences in Kalisz, Nowy Šwiat 4, 62-800 Kalisz, Poland
| | - M Wojciechowska
- Mother and Child Health Department, Karol Marcinkowski Medical University in Poznań, ul. Fredry 10, 61-701 Poznan, Poland
| | - Z Chęcińska-Maciejewska
- Physiology Department, Karol Marcinkowski Medical University in Poznań, ul. Fredry 10, 61-701 Poznan, Poland
| | - P Kaczmarek
- Department of Medicine, The President Stanisław Wojciechowski State University of Applied Sciences in Kalisz, Nowy Šwiat 4, 62-800 Kalisz, Poland
| | - H Sommermeyer
- Department of Medicine, The President Stanisław Wojciechowski State University of Applied Sciences in Kalisz, Nowy Šwiat 4, 62-800 Kalisz, Poland
| |
Collapse
|
49
|
Simonson J, Haglund K, Weber E, Fial A, Hanson L. Probiotics for the Management of Infantile Colic: A Systematic Review. MCN Am J Matern Child Nurs 2021; 46:88-96. [PMID: 33315632 DOI: 10.1097/nmc.0000000000000691] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
BACKGROUND Colic is defined as periods of inconsolable crying, fussing, or irritability that have no apparent cause and present in healthy infants under 5 months of age. Although colic is a benign and self-limiting condition, it can be distressing to parents and there are few robust treatment interventions. This systematic review explored the evidence for administration of probiotics to prevent or decrease symptoms of colic. METHODS Literature searches were conducted in PubMed, CINAHL (Cumulative Index to Nursing and Allied Health Literature), Cochrane Library, and Web of Science. SAMPLE Twenty articles were included: 15 randomized controlled trials and 5 meta-analyses. RESULTS Based on the evidence in this systematic review, the oral administration of probiotics to breastfed infants with colic resulted in at least a 50% reduction in crying time compared with placebo. Efficacy of probiotics to reduce colic symptoms in formula-fed infants needs further study. In this review, we did not find evidence to support or refute efficacy of probiotics to prevent infantile colic. Clinical Implication: Probiotics (especially the strain Lactobacillus reuteri DSM 17938) can safely be recommended if parents desire a treatment option for their infants with colic.
Collapse
|
50
|
Chen K, Liu C, Li H, Lei Y, Zeng C, Xu S, Li J, Savino F. Infantile Colic Treated With Bifidobacterium longum CECT7894 and Pediococcus pentosaceus CECT8330: A Randomized, Double-Blind, Placebo-Controlled Trial. Front Pediatr 2021; 9:635176. [PMID: 34568236 PMCID: PMC8461252 DOI: 10.3389/fped.2021.635176] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 08/12/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Colic is a common condition in infants <4 months of age. Attempts to treat infantile colic with probiotics have shown variable efficacy and overall low evidence of success. In this work, we tested the hypothesis that oral administration of Bifidobacterium longum CECT7894 (KABP042) and Pediococcus pentosaceus CECT8330 (KABP041) mix (1 × 109 colony forming units) would improve the symptoms of infantile colic. Methods: A total of 112 exclusively breastfed or mixed fed infants aged <2 months and meeting the ROME IV criteria for infantile colic were recruited. The infants were randomized in a double-blind, placebo-controlled trial to receive orally administered probiotics (intervention group, IG, n = 48) or placebo (placebo group, PG, n = 42) daily for 21 days. Results: Infants in the IG had significantly shorter crying time (p < 0.001) on day 7 [IG vs. PG, median (25-75th percentile): 38 (3.5-40.5) vs. 62 (40-108) min/day], day 14 [IG vs. PG: 20 (0-40) vs. 50 (30-75) min/day], and day 21 [IG vs. PG: 14 (0-33) vs. 40 (28-62) min/day]. Higher responder ratio and fewer crying/fussing episodes on days 7, 14, and 21 and better stool consistency on day 21 were observed in the IG (p < 0.01) as compared to the PG. Conversely, no significant effects on stool frequency or quality of life were observed. Conclusions: In summary, daily oral administration of B. longum CECT7894 (KABP042) and P. pentosaceus CECT8330 (KABP041) was an effective treatment for shortening crying time due to infantile colic and for improving fecal consistency. This trial was registered retrospectively in December 2019 with a trial number of ISRCTN92431452.
Collapse
Affiliation(s)
- Ke Chen
- Department of Nutrition, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.,Department of Child Health Care, Angel Children's Hospital, Chengdu, China
| | - Changqi Liu
- School of Exercise and Nutritional Sciences, San Diego State University, San Diego, CA, United States
| | - Hua Li
- Department of Child Health Care, Qingbaijiang Maternal and Child Health Hospital, Chengdu, China
| | - Yuehua Lei
- Department of Child Health Care, Qingbaijiang Maternal and Child Health Hospital, Chengdu, China
| | - Chenggui Zeng
- Department of Child Health Care, Chengdu Caojiaxiang Community Healthcare Center, Chengdu, China
| | - Shuhong Xu
- Department of Child Health Care, Huili Maternity and Child Care Center, Huili, China
| | - Jianqiu Li
- Department of Child Health Care, Angel Children's Hospital, Chengdu, China
| | - Francesco Savino
- Department of Paediatrics, S.S.D. Subintensive Neonatal Care, Children Hospital 'Regina Margherita', Turin, Italy
| |
Collapse
|