1
|
He Y, Zhang B, Xin Y, Wang W, Wang X, Liu Z, She Y, Guo R, Jia G, Wu S, Liu Z. Synbiotic combination of 2'-fucosyllactose and Bifidobacterium mitigates neurodevelopmental disorders and ASD-like behaviors induced by valproic acid. Food Funct 2025; 16:2703-2717. [PMID: 40165714 DOI: 10.1039/d4fo06234e] [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: 04/02/2025]
Abstract
Autism spectrum disorder (ASD) is a group of heterogeneous neurodevelopmental disorders characterized by social deficits and repetitive behaviors. Increasing evidence suggests that the gut microbiota influences neurodevelopment and behavior. In this study, we established an ASD model by administering valproic acid (VPA) to pregnant females, with male offspring receiving a daily synbiotic intervention for four weeks post-weaning. The results indicate that the synbiotic combination of 2'-Fucosyllactose (2'-FL) and Bifidobacterium animalis subsp. lactis BB-12 (BB-12) outperformed that of 2'-FL and Lactobacillus paracasei L9300BH (L9300BH) in alleviating social deficits, repetitive behaviors, neuronal damage, and dysregulated expression of social-related genes and neuroinflammatory markers in ASD mice. Additionally, the intervention with 2'-FL and BB-12 improved gut morphology and barrier integrity, reduced gut inflammation, and optimized the gut microbiota structure by increasing the abundance of Bifidobacteria and Akkermansia, known producers of short-chain fatty acids (SCFAs). Notably, levels of acetate, propionate, and butyrate were significantly elevated in fecal samples. In summary, the synbiotic combination of 2'-FL and BB-12 supports gut microbiota homeostasis, enhances fecal SCFA levels, and mitigates neurodevelopmental abnormalities in ASD mice.
Collapse
Affiliation(s)
- Ying He
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China.
| | - Bo Zhang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China.
| | - Yu Xin
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China.
| | - Wenxiu Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China.
| | - Xue Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China.
| | - Zhuo Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China.
- Research and Development Center, Xi'an Yinqiao Dairy (Group) Co., Ltd, Xi'an, Shaanxi 710075, China
| | - Yongbo She
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China.
| | - Rui Guo
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China.
- Northwest A&F University Shenzhen Research Institute, Shenzhen, Guangdong, 518000, China
| | - Gengjie Jia
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, China
| | - Shan Wu
- Research and Development Center, Xi'an Yinqiao Dairy (Group) Co., Ltd, Xi'an, Shaanxi 710075, China
| | - Zhigang Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China.
- Northwest A&F University Shenzhen Research Institute, Shenzhen, Guangdong, 518000, China
- Shaanxi Precision Nutrition and Health Research Institute, Xian 710300, China
| |
Collapse
|
2
|
Sujaya IN, Mariyatun M, Hasan PN, Manurung NEP, Pramesi PC, Juffrie M, Utami T, Cahyanto MN, Yamamoto S, Takahashi T, Asahara T, Akiyama T, Rahayu ES. Randomized study of Lacticaseibacillus fermented milk in Indonesian elderly houses: Impact on gut microbiota and gut environment. World J Gastroenterol 2025; 31:104081. [PMID: 40182598 PMCID: PMC11962840 DOI: 10.3748/wjg.v31.i12.104081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/21/2025] [Accepted: 02/24/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Health maintenance in elderly houses includes management of the gut microbiota and the environment. Lacticaseibacillus paracasei Shirota (LcS) is a probiotic strain that positively affects the human gut. However, the evidence of its effects on the Indonesian population remains limited. AIM To investigate the effect of LcS-fermented milk on the gut microbiota and environment of Indonesian elderly houses. METHODS This double-blind, randomized, placebo-controlled trial involved 112 participants from Indonesian elderly houses, spanning a 2-week baseline and 24-week treatment. Participants were randomly assigned to probiotic or placebo groups, consuming fermented milk with or without LcS (> 6.5 × 109 colony-forming units). Fecal samples were collected every three months. Gut microbiota analysis was performed using 16S rRNA gene sequencing and reverse transcription quantitative polymerase chain reaction, while gut environment was assessed by measuring fecal organic acids, amino acid metabolites, and stool frequency. RESULTS Analyses of 16S rRNA gene sequence data at the 3-month period revealed increased Bifidobacterium and Succinivibrio and decreased Rikenellaceae RC9 gut group in the probiotic group. These shifts were associated with significant differences in β-diversity metrics. The change in Bifidobacterium was confirmed by reverse transcription quantitative polymerase chain reaction, demonstrating higher abundance in the probiotic group than in the placebo group (8.5 ± 1.1 vs 8.0 ± 1.1, log10 bacterial cells/g; P = 0.044). At 6-month period, the differences in Succinivibrio and Rikenellaceae RC9 gut group persisted. The probiotic group showed higher butyrate levels than the placebo group at the 6-month period (5.04 ± 3.11 vs 3.95 ± 2.89, μmol/g; P = 0.048). The effect on amino acid metabolites and stool frequency was not significant. CONCLUSION Daily intake of LcS positively affects the gut microbiota and environment of people living in Indonesian elderly houses.
Collapse
Affiliation(s)
- I Nengah Sujaya
- School of Public Health, Faculty of Medicine, Udayana University, Denpasar 80230, Bali, Indonesia
| | - Mariyatun Mariyatun
- Center for Food and Nutrition Studies, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
- Center of Excellence for Research and Application on Integrated Probiotics Industry, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
| | - Pratama Nur Hasan
- Center for Food and Nutrition Studies, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
- Center of Excellence for Research and Application on Integrated Probiotics Industry, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
| | - Nancy Eka Putri Manurung
- Center for Food and Nutrition Studies, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
- Center of Excellence for Research and Application on Integrated Probiotics Industry, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
| | - Putrika Citta Pramesi
- Center for Food and Nutrition Studies, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
- Center of Excellence for Research and Application on Integrated Probiotics Industry, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
- Department of Food and Agricultural Product Technology, Faculty of Agricultural Technology, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
| | - Mohammad Juffrie
- Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
| | - Tyas Utami
- Center of Excellence for Research and Application on Integrated Probiotics Industry, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
- Department of Food and Agricultural Product Technology, Faculty of Agricultural Technology, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
| | - Muhammad Nur Cahyanto
- Department of Food and Agricultural Product Technology, Faculty of Agricultural Technology, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
| | - Shuta Yamamoto
- Yakult Central Institute, Yakult Honsha Co., Ltd., Kunitachi 186-8650, Tōkyō, Japan
| | - Takuya Takahashi
- Yakult Honsha European Research Center for Microbiology VOF, Ghent 9052, East Flanders, Belgium
| | - Takashi Asahara
- Yakult Central Institute, Yakult Honsha Co., Ltd., Kunitachi 186-8650, Tōkyō, Japan
| | - Takuya Akiyama
- Yakult Central Institute, Yakult Honsha Co., Ltd., Kunitachi 186-8650, Tōkyō, Japan
| | - Endang Sutriswati Rahayu
- Center for Food and Nutrition Studies, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
- Center of Excellence for Research and Application on Integrated Probiotics Industry, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
- Department of Food and Agricultural Product Technology, Faculty of Agricultural Technology, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
| |
Collapse
|
3
|
Sawane K, Takahashi I, Ishikuro M, Takumi H, Orui M, Noda A, Shinoda G, Ohseto H, Onuma T, Ueno F, Murakami K, Higuchi N, Tanaka T, Furuyashiki T, Nakamura T, Koshiba S, Ohneda K, Kumada K, Ogishima S, Hozawa A, Sugawara J, Kuriyama S, Obara T. Association Between Human Milk Oligosaccharides and Early Adiposity Rebound in Children: A Case-Control Study of the Tohoku Medical Megabank Project Birth and Three-Generation Cohort Study. J Nutr 2025:S0022-3166(25)00150-6. [PMID: 40058699 DOI: 10.1016/j.tjnut.2025.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 01/15/2025] [Accepted: 02/11/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Adiposity rebound (AR) is the point when the BMI begins to rise again during early childhood. Early AR (before age 5) is associated with higher risk of lifelong obesity and metabolic disorders and may be influenced by breastfeeding. Although human milk oligosaccharides (HMOs) in breast milk are crucial for child growth, their association with AR status has not been studied. OBJECTIVES This study aimed to explore the association between breast milk HMOs and AR status in children. METHODS In this case-control study, we included 184 mother-child pairs from the Tohoku Medical Megabank Project Birth and Three-Generation (TMM BirThree) Cohort Study (93 AR cases, 91 controls). Breast milk was collected 1 mo postpartum, and the concentration of 15 HMO molecules and α-diversity index (Inverse Simpson index) were quantified. Wilcoxon rank-sum test and partial least squares-discriminant analysis identified candidate HMOs, and multivariable logistic regression analysis evaluated associations between candidate HMOs and AR status. Analyses were stratified by maternal secretor status (secretor or nonsecretor). RESULTS In secretor mothers, multivariable logistic regression showed that the inverse Simpson index [odds ratio (OR): 0.54; 95% CI: 0.36, 0.82), the sum of sialic acid-bound HMOs (OR: 0.61; 95% CI: 0.41, 0.91), and 3'-sialyllactose (OR: 0.67; 95% CI: 0.46, 0.98) were inversely associated with early AR in the fully adjusted model. A trend of interaction between sialyl-lacto-N-tetraose-a (LSTa) and maternal secretor status regarding AR was observed in the fully adjusted model (P-interaction = 0.051). CONCLUSIONS α-Diversity, sialic acid-bound HMOs, and 3'-sialyllactose may involved in inhibiting AR in children of secretor mothers, and a trend of interactive effect between LSTa and maternal secretor status regarding AR is indicated. These findings offer novel perspectives on the associations between breastfeeding and a childhood adiposity as well as potential metabolic disorders later in life. This trial is registered at https://www.umin.ac.jp/ as UMIN000047160.
Collapse
Affiliation(s)
| | - Ippei Takahashi
- Division of Molecular Epidemiology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Mami Ishikuro
- Division of Molecular Epidemiology, Graduate School of Medicine, Tohoku University, Sendai, Japan; Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | | | - Masatsugu Orui
- Division of Molecular Epidemiology, Graduate School of Medicine, Tohoku University, Sendai, Japan; Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Aoi Noda
- Division of Molecular Epidemiology, Graduate School of Medicine, Tohoku University, Sendai, Japan; Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan; Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan
| | - Genki Shinoda
- Division of Molecular Epidemiology, Graduate School of Medicine, Tohoku University, Sendai, Japan; Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Hisashi Ohseto
- Division of Molecular Epidemiology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Tomomi Onuma
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Fumihiko Ueno
- Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Keiko Murakami
- Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | | | | | | | - Tomohiro Nakamura
- Department of Health Record Informatics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan; Faculty of Data Science, Kyoto Women's University, Kyoto, Japan
| | - Seizo Koshiba
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan; Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Sendai, Japan
| | - Kinuko Ohneda
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Sendai, Japan; Department of Biobank, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Kazuki Kumada
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Sendai, Japan; Department of Biobank, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Soichi Ogishima
- Department of Health Record Informatics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan; Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Sendai, Japan
| | - Atsushi Hozawa
- Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan; Division of Epidemiology, School of Public Health, Graduate School of Medicine, Tohoku University, Sendai, Japan; Division of Personalized Prevention and Epidemiology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | | | - Shinichi Kuriyama
- Division of Molecular Epidemiology, Graduate School of Medicine, Tohoku University, Sendai, Japan; Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan; Division of Disaster Public Health, International Research Institute of Disaster Science, Tohoku University, Sendai, Japan
| | - Taku Obara
- Division of Molecular Epidemiology, Graduate School of Medicine, Tohoku University, Sendai, Japan; Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan; Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan.
| |
Collapse
|
4
|
Yassin LK, Nakhal MM, Alderei A, Almehairbi A, Mydeen AB, Akour A, Hamad MIK. Exploring the microbiota-gut-brain axis: impact on brain structure and function. Front Neuroanat 2025; 19:1504065. [PMID: 40012737 PMCID: PMC11860919 DOI: 10.3389/fnana.2025.1504065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/30/2025] [Indexed: 02/28/2025] Open
Abstract
The microbiota-gut-brain axis (MGBA) plays a significant role in the maintenance of brain structure and function. The MGBA serves as a conduit between the CNS and the ENS, facilitating communication between the emotional and cognitive centers of the brain via diverse pathways. In the initial stages of this review, we will examine the way how MGBA affects neurogenesis, neuronal dendritic morphology, axonal myelination, microglia structure, brain blood barrier (BBB) structure and permeability, and synaptic structure. Furthermore, we will review the potential mechanistic pathways of neuroplasticity through MGBA influence. The short-chain fatty acids (SCFAs) play a pivotal role in the MGBA, where they can modify the BBB. We will therefore discuss how SCFAs can influence microglia, neuronal, and astrocyte function, as well as their role in brain disorders such as Alzheimer's disease (AD), and Parkinson's disease (PD). Subsequently, we will examine the technical strategies employed to study MGBA interactions, including using germ-free (GF) animals, probiotics, fecal microbiota transplantation (FMT), and antibiotics-induced dysbiosis. Finally, we will examine how particular bacterial strains can affect brain structure and function. By gaining a deeper understanding of the MGBA, it may be possible to facilitate research into microbial-based pharmacological interventions and therapeutic strategies for neurological diseases.
Collapse
Affiliation(s)
- Lidya K. Yassin
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mohammed M. Nakhal
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Alreem Alderei
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Afra Almehairbi
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Ayishal B. Mydeen
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Amal Akour
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mohammad I. K. Hamad
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
5
|
Cai R, Zhang J, Song Y, Liu X, Xu H. Research Progress on the Degradation of Human Milk Oligosaccharides (HMOs) by Bifidobacteria. Nutrients 2025; 17:519. [PMID: 39940377 PMCID: PMC11820314 DOI: 10.3390/nu17030519] [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: 01/03/2025] [Revised: 01/26/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025] Open
Abstract
The purpose of this study was to investigate the degradation mechanism of Bifidobacterium on breast milk oligosaccharides (HMOs) and its application in infant nutrition. The composition and characteristics of HMOs were introduced, and the degradation mechanism of HMOs by Bifidobacterium was described, including intracellular and extracellular digestion and species-specific differences. The interaction between Bifidobacterium and Bacteroides in the process of degrading HMOs and its effect on intestinal microecology were analyzed. The effects of HMO formula milk powder on the intestinal microbiota of infants were discussed, including simulating breast milk composition, regulating intestinal flora and immune function, infection prevention, and brain development. Finally, the research results are summarized, and future research directions are proposed to provide directions for research in the field of infant nutrition.
Collapse
Affiliation(s)
| | | | | | - Xiaoyong Liu
- School of Biological Science and Technology, University of Jinan, Jinan 250024, China; (R.C.); (J.Z.); (Y.S.)
| | - Huilian Xu
- School of Biological Science and Technology, University of Jinan, Jinan 250024, China; (R.C.); (J.Z.); (Y.S.)
| |
Collapse
|
6
|
Sun W, Tao L, Qian C, Xue PP, Du SS, Tao YN. Human milk oligosaccharides: bridging the gap in intestinal microbiota between mothers and infants. Front Cell Infect Microbiol 2025; 14:1386421. [PMID: 39835278 PMCID: PMC11743518 DOI: 10.3389/fcimb.2024.1386421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 12/04/2024] [Indexed: 01/22/2025] Open
Abstract
Breast milk is an essential source of infant nutrition. It is also a vital determinant of the structure and function of the infant intestinal microbial community, and it connects the mother and infant intestinal microbiota. Human milk oligosaccharides (HMOs) are a critical component in breast milk. HMOs can reach the baby's colon entirely from milk and become a fermentable substrate for some intestinal microorganisms. HMOs can enhance intestinal mucosal barrier function and affect the intestinal function of the host through immune function, which has a therapeutic effect on specific infant intestinal diseases, such as necrotizing enterocolitis. In addition, changes in infant intestinal microbiota can reflect the maternal intestinal microbiota. HMOs are a link between the maternal intestinal microbiota and infant intestinal microbiota. HMOs affect the intestinal microbiota of infants and are related to the maternal milk microbiota. Through breastfeeding, maternal microbiota and HMOs jointly affect infant intestinal bacteria. Therefore, HMOs positively influence the establishment and balance of the infant microbial community, which is vital to ensure infant intestinal function. Therefore, HMOs can be used as a supplement and alternative therapy for infant intestinal diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Ying-na Tao
- Department of Traditional Chinese Medicine, Shanghai Fourth People’s Hospital
Affiliated to Tongji University, Shanghai, China
| |
Collapse
|
7
|
Du Q, Liu X, Zhang R, Hu G, Liu Q, Wang R, Ma W, Hu Y, Fan Z, Li J. Placental and Fetal Microbiota in Rhesus Macaque: A Case Study Using Metagenomic Sequencing. Am J Primatol 2025; 87:e23718. [PMID: 39716039 DOI: 10.1002/ajp.23718] [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: 08/26/2024] [Revised: 11/04/2024] [Accepted: 11/30/2024] [Indexed: 12/25/2024]
Abstract
Recent evidence challenging the notion of a sterile intrauterine environment has sparked research into the origins and effects of fetal microbiota on immunity development during gestation. Rhesus macaques (RMs) serve as valuable nonhuman primate models due to their similarities to humans in development, placental structure, and immune response. In this study, metagenomic analysis was applied to the placenta, umbilical cord, spleen, gastrointestinal tissues of an unborn RM fetus, and the maternal intestine, revealing the diversity and functionality of microbes in these tissues. Additionally, gut metagenomic data of adult Rhesus macaques from our previous study, along with data from a human fetus obtained from public databases, were included for comparison. We observed substantial microbial sharing between the mother and fetus, with the microbial composition of the placenta and umbilical cord more closely resembling that of the fetal organs than the maternal intestine. Notably, compared with other adult RMs, there was a clear convergence between maternal and fetal microbiota, alongside distinct differences between the microbiota of adults and the fetus, which underscores the unique microbial profiles in fetal environments. Furthermore, the fetal microbiota displayed a less developed carbohydrate metabolism capacity than adult RMs. It also shared antibiotic resistance genes with both maternal and adult RM microbiomes, indicating potential vertical transmission. Comparative analysis of the metagenomes between the RM fetus and a human fetus revealed significant differences in microbial composition and genes, yet also showed similarities in certain abundant microbiota. Collectively, our results contribute to a more comprehensive understanding of the intrauterine microbial environment in macaques.
Collapse
Affiliation(s)
- Qiao Du
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, China
| | - Xu Liu
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, China
| | - Rusong Zhang
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, China
| | - Gang Hu
- SCU-SGHB Joint Laboratory on Nonhuman Primates Research, Sichuan Green-house Biotech Co. Ltd., Meishan, China
| | - Qinghua Liu
- SCU-SGHB Joint Laboratory on Nonhuman Primates Research, Sichuan Green-house Biotech Co. Ltd., Meishan, China
| | - Rui Wang
- SCU-SGHB Joint Laboratory on Nonhuman Primates Research, Sichuan Green-house Biotech Co. Ltd., Meishan, China
| | - Wen Ma
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, China
| | - Ying Hu
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, China
| | - Zhenxin Fan
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, China
| | - Jing Li
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, China
| |
Collapse
|
8
|
Duman H, Bechelany M, Karav S. Human Milk Oligosaccharides: Decoding Their Structural Variability, Health Benefits, and the Evolution of Infant Nutrition. Nutrients 2024; 17:118. [PMID: 39796552 PMCID: PMC11723173 DOI: 10.3390/nu17010118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 12/24/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025] Open
Abstract
Human milk oligosaccharides (HMOs), the third most abundant solid component in human milk, vary significantly among women due to factors such as secretor status, race, geography, season, maternal nutrition and weight, gestational age, and delivery method. In recent studies, HMOs have been shown to have a variety of functional roles in the development of infants. Because HMOs are not digested by infants, they act as metabolic substrates for certain bacteria, helping to establish the infant's gut microbiota. By encouraging the growth of advantageous intestinal bacteria, these sugars function as prebiotics and produce short-chain fatty acids (SCFAs), which are essential for gut health. HMOs can also specifically reduce harmful microbes and viruses binding to the gut epithelium, preventing illness. HMO addition to infant formula is safe and promotes healthy development, infection prevention, and microbiota. Current infant formulas frequently contain oligosaccharides (OSs) that differ structurally from those found in human milk, making it unlikely that they would reproduce the unique effects of HMOs. However, there is a growing trend in producing OSs resembling HMOs, but limited data make it unclear whether HMOs offer additional therapeutic benefits compared to non-human OSs. Better knowledge of how the human mammary gland synthesizes HMOs could direct the development of technologies that yield a broad variety of complex HMOs with OS compositions that closely mimic human milk. This review explores HMOs' complex nature and vital role in infant health, examining maternal variation in HMO composition and its contributing factors. It highlights recent technological advances enabling large-scale studies on HMO composition and its effects on infant health. Furthermore, HMOs' multifunctional roles in biological processes such as infection prevention, brain development, and gut microbiota and immune response regulation are investigated. The structural distinctions between HMOs and other mammalian OSs in infant formulas are discussed, with a focus on the trend toward producing more precise replicas of HMOs found in human milk.
Collapse
Affiliation(s)
- Hatice Duman
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17100, Türkiye;
| | - Mikhael Bechelany
- Institut Européen des Membranes (IEM), UMR 5635, University Montpellier, ENSCM, CNRS, F-34095 Montpellier, France
- Functional Materials Group, Gulf University for Science and Technology (GUST), Masjid Al Aqsa Street, Mubarak Al-Abdullah 32093, Kuwait
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17100, Türkiye;
| |
Collapse
|
9
|
Mady EA, Osuga H, Toyama H, El-Husseiny HM, Inoue R, Murase H, Yamamoto Y, Nagaoka K. Relationship between the components of mare breast milk and foal gut microbiome: shaping gut microbiome development after birth. Vet Q 2024; 44:1-9. [PMID: 38733121 PMCID: PMC11089936 DOI: 10.1080/01652176.2024.2349948] [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: 09/09/2023] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
The gut microbiota (GM) is essential for mammalian health. Although the association between infant GM and breast milk (BM) composition has been well established in humans, such a relationship has not been investigated in horses. Hence, this study was conducted to analyze the GM formation of foals during lactation and determine the presence of low-molecular-weight metabolites in mares' BM and their role in shaping foals' GM. The fecal and BM samples from six pairs of foals and mares were subjected to 16S ribosomal RNA metagenomic and metabolomic analyses, respectively. The composition of foal GM changed during lactation time; hierarchical cluster analysis divided the fetal GM into three groups corresponding to different time points in foal development. The level of most metabolites in milk decreased over time with increasing milk yield, while threonic acid and ascorbic acid increased. Further analyses revealed gut bacteria that correlated with changes in milk metabolites; for instance, there was a positive correlation between Bacteroidaceae in the foal's gut microbiota and serine/glycine in the mother's milk. These findings help improve the rearing environment of lactating horses and establish artificial feeding methods for foals.
Collapse
Affiliation(s)
- Eman A. Mady
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
- Department of Animal Hygiene, Behavior, and Management, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Elqaliobiya, Egypt
| | - Haruna Osuga
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Haruka Toyama
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Hussein M. El-Husseiny
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Ryo Inoue
- Laboratory of Animal Science, Department of Applied Biological Science, Setsunan University, Osaka, Japan
| | - Harutaka Murase
- Hidaka Training and Research Center, Japan Racing Association, Hokkaido, Japan
| | - Yuki Yamamoto
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Kentaro Nagaoka
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| |
Collapse
|
10
|
Biagioli V, Sortino V, Falsaperla R, Striano P. Role of Human Milk Microbiota in Infant Neurodevelopment: Mechanisms and Clinical Implications. CHILDREN (BASEL, SWITZERLAND) 2024; 11:1476. [PMID: 39767905 PMCID: PMC11674883 DOI: 10.3390/children11121476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 11/23/2024] [Accepted: 11/29/2024] [Indexed: 01/05/2025]
Abstract
BACKGROUND Human milk (HM) is recognized as an ideal source of nutrition for newborns; as a result, its multiple bioactive molecules can support the growth of healthy newborns and reduce the risk of mortality and diseases such as asthma, respiratory infections, diabetes (type 1 and 2), and gastrointestinal disorders such as ulcerative colitis and Crohn's disease. Furthermore, it can reduce the severity of necrotizing enterocolitis (NEC) in preterm infants. Moreover, human milk oligosaccharides (HMOs) present in breast milk show an immunomodulatory, prebiotic, and neurodevelopmental effect that supports the microbiota-gut-brain axis. MATERIAL AND METHODS This study examined the state-of-the-art research, using keywords such as "breastfeeding", "human milk oligosaccharides", "microbiota-gut-brain axis", "infants", and "malnutrition". The literature review was conducted by selecting articles between 2013 and 2024, as the most recent ones. The databases used were Web Science, PubMed, and Scopus. RESULTS We found multiple studies examining the composition of HM and infant formula (IF). However, further longitudinal studies and randomized control trials (RCTs) are needed to better understand the clinical outcomes that bioactive components exert on healthy and hospitalized children and how, in conditions of malnutrition, it is necessary to support the growth of the newborn. CONCLUSIONS In this review, we affirm the importance of human milk and, through it, the modulation of the microbiota and the neuroprotective role in newborns, determining the health of the following years of life.
Collapse
Affiliation(s)
- Valentina Biagioli
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16126 Genoa, Italy;
| | - Vincenzo Sortino
- Unit of Pediatrics and Pediatric Emergency, Azienda Ospedaliero-Universitaria Policlinico “Rodolico-San Marco”, San Marco Hospital, University of Catania, 95123 Catania, Italy;
| | - Raffaele Falsaperla
- Department of Medical Science-Pediatrics, University of Ferrara, 44124 Ferrara, Italy;
| | - Pasquale Striano
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16126 Genoa, Italy;
- Pediatric Neurology and Neuromuscular Diseases Unit, IRCCS Giannina Gaslini Full Member of EPICARE, 16121-16167 Genoa, Italy
| |
Collapse
|
11
|
Kawikova I, Hakenova K, Lebedeva M, Kleteckova L, Jakob L, Spicka V, Wen L, Spaniel F, Vales K. Perinatal Hypoxia and Immune System Activation in Schizophrenia Pathogenesis: Critical Considerations During COVID-19 Pandemic. Physiol Res 2024; 73:S615-S639. [PMID: 39589306 PMCID: PMC11627263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 10/01/2024] [Indexed: 11/27/2024] Open
Abstract
Schizophrenia, a severe psychiatric, neurodevelopmental disorder affecting about 0.29-1 % of the global population, is characterized by hallucinations, delusions, cognitive impairments, disorganized thoughts and speech, leading to significant social withdrawal and emotional blunting. During the 1980s, considerations about diseases that result from complex interactions of genetic background and environmental factors started to appear. One of the critical times of vulnerability is the perinatal period. Concerning schizophrenia, obstetric complications that are associated with hypoxia of the fetus or neonate were identified as a risk. Also, maternal infections during pregnancy were linked to schizophrenia by epidemiological, serologic and genetic studies. Research efforts then led to the development of experimental models testing the impact of perinatal hypoxia or maternal immune activation on neurodevelopmental disorders. These perinatal factors are usually studied separately, but given that the models are now validated, it is feasible to investigate both factors together. Inclusion of additional factors, such as metabolic disturbances or chronic stress, may need to be considered also. Understanding the interplay of perinatal factors in schizophrenia's etiology is crucial for developing targeted prevention and therapeutic strategies.
Collapse
Affiliation(s)
- I Kawikova
- Department of Medicine, Yale University, New Haven, CT, USA,
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Li A, Kou R, Wang J, Zhang B, Zhang Y, Liu J, Hu Y, Wang S. 2'-Fucosyllactose ameliorates aging-related osteoporosis by restoring gut microbial and innate immune homeostasis. J Adv Res 2024:S2090-1232(24)00536-8. [PMID: 39550028 DOI: 10.1016/j.jare.2024.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 10/04/2024] [Accepted: 11/11/2024] [Indexed: 11/18/2024] Open
Abstract
INTRODUCTION Aging-related osteoporosis is considered as a serious public health concern for middle-aged and elderly people, with an intricated pathogenesis including the recently identified aging-induced immunological dysfunction and gut microbial disorder. The intervention based on dietary prebiotics is recommended to retain bone health and postpone the progression of osteoporosis. OBJECTIVES As a well-defined prebiotic, 2'-fucosyllactose (2'-FL) has been thoroughly validated with positive effect on systemic health and was proposed in this study to unveil its intervention on aging-related osteoporosis, as well as the underlying mechanisms involving the gut microecology and innate immunity. METHODS The effects of dietary 2'-FL on osteoporosis phenotypes were identified by evaluating the severity of bone loss and microstructure damage in natural aging mice. The mechanisms relying on innate immune profile, intestinal barrier function, and gut microbial homeostasis, were analyzed to elucidate the signaling axis. The detailed molecular signaling was validated based on LPS-stimulated RAW 264.7 murine macrophages. RESULTS The results indicated that 12-week 2'-FL intervention retrieved bone loss and microstructure damage in natural aging mice. Also, 2'-FL alleviated aging-induced colonic inflammation, gut barrier dysfunction, and abnormal expression of intestinal tight-junction protein. The impact of 2'-FL treatment on the aging-induced gut microbial dysbiosis was validated by restoring gut microbiota diversity, recovering the abundance of Bifidobacterium, Prevotellaceae and Akkermansia, and inhibiting the growth of Stenotrophomonas. Flow cytometry analysis revealed changes in dendritic cell (DC) and macrophage subsets with age, and a decrease in M1-polarized macrophages was observed in 2'-FL-treated aged mice and RAW264.7 cells potentially through the interaction with toll-like receptor 4 (TLR4) to suppress NF-κB signaling and the secretion of proinflammatory factors. CONCLUSION These findings highlight the preventive effect of 2'-FL on aging-associated osteoporosis by regulating gut microbial homeostasis and innate immune responses.
Collapse
Affiliation(s)
- Ang Li
- School of Medicine, Nankai University, Tianjin, China
| | - Ruixin Kou
- School of Medicine, Nankai University, Tianjin, China
| | - Jin Wang
- School of Medicine, Nankai University, Tianjin, China
| | - Bowei Zhang
- School of Medicine, Nankai University, Tianjin, China
| | - Yan Zhang
- School of Medicine, Nankai University, Tianjin, China
| | - Jingmin Liu
- School of Medicine, Nankai University, Tianjin, China
| | - Yaozhong Hu
- School of Medicine, Nankai University, Tianjin, China.
| | - Shuo Wang
- School of Medicine, Nankai University, Tianjin, China.
| |
Collapse
|
13
|
Jiang S, Qin J, Shi L, Feng J, Mo J, Su W, Cheng Y, Lv J, Li Q, Li S, Zeng L, Han B, Zhou J. Association among Gestational Weight Gain, Fucosylated Human Milk Oligosaccharides, and Breast Milk Microbiota─An Evidence in Healthy Mothers from Northwest China. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:25135-25145. [PMID: 39476856 PMCID: PMC11565758 DOI: 10.1021/acs.jafc.4c07050] [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: 08/03/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 11/14/2024]
Abstract
This study investigates the relationship among maternal secretor status, human milk oligosaccharides (HMOs), and the composition of breastmilk microbiota in a cohort of healthy mothers from Shaanxi province, China. The results demonstrated that 78.9% of the mothers were secretors, exhibiting an active fucosyltransferase 2 gene (fut2) and producing α-1,2 fucosylated HMOs, which significantly affected the HMO profile. Secretor mothers had higher levels of 2'-FL and LNFPI in contrast to nonsecretors who displayed high levels of 3'-FL, LNFPII, and LNT. Furthermore, secretor mothers exhibited greater diversity in HMOs compared with nonsecretors, although no significant differences were observed in the breast milk microbiota composition. A correlation was identified between specific HMOs (2'-FL, 3'-FL, 6'-SL, and LNFPI) and the microbiota composition. Notably, mothers with normal weight gain during pregnancy demonstrated higher microbial diversity, with increased abundance of beneficial genera such as Bifidobacterium, Lactobacillus, and Ligilactobacillus. These findings contribute to the development of potential guidelines for providing personalized nutrition.
Collapse
Affiliation(s)
- Sijin Jiang
- School
of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Jiale Qin
- School
of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Lu Shi
- School
of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Jiayu Feng
- School
of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Jianhui Mo
- School
of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Wanghong Su
- School
of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Yue Cheng
- School
of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Jia Lv
- School
of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
- Department
of Pediatrics, The Second Affiliated Hospital
of Xi’an Jiaotong University, Xi’an, Shaanxi 710004, China
| | - Qiang Li
- School
of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Shaoru Li
- School
of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Lingxia Zeng
- School
of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Bei Han
- School
of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Jing Zhou
- Department
of Pediatrics, The Second Affiliated Hospital
of Xi’an Jiaotong University, Xi’an, Shaanxi 710004, China
| |
Collapse
|
14
|
Long J, Zhang J, Zeng X, Wang M, Wang N. Prevention and Treatment of Alzheimer's Disease Via the Regulation of the Gut Microbiota With Traditional Chinese Medicine. CNS Neurosci Ther 2024; 30:e70101. [PMID: 39508315 PMCID: PMC11541599 DOI: 10.1111/cns.70101] [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: 05/29/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 11/15/2024] Open
Abstract
Alzheimer's disease (AD) is caused by a variety of factors, and one of the most important factors is gut microbiota dysbiosis. An imbalance in the gut mincrobiota have been shown to change the concentrations of lipopolysaccharide and short-chain fatty acids. These microorganisms synthesize substances that can influence the levels of a variety of metabolites and cause multiple diseases through the immune response, fatty acid metabolism, and amino acid metabolism pathways. Furthermore, these metabolic changes promote the formation of β-amyloid plaques and neurofibrillary tangles. Thus, the microbiota-gut-brain axis plays an important role in AD development. In addition to traditional therapeutic drugs such as donepezil and memantine, traditional Chinese medicines (TCMs) have also showed to significantly decrease the severity of AD symptoms and suppress the underlying related mechanisms. We searched for studies on the effects of different herbal monomers, single herbs, and polyherbal formulas on the gut microbiota of AD patients and identified the relevant pathways through which the gut microbiota affected AD. We conclude that improvements in the gut microbiota not only decrease the occurrence of inflammatory reactions but also reduce the deposition of central pathological products. Herbal monomers have a stronger effect on improving of central pathology. Polyherbal formulas have the most extensive effect on the gut microbiota in patients with AD. Among the effects of formulas, the anti-inflammatory effect is the most essential and is also the main concern regarding the use of TCMs in treating AD from the viewpoint of the gut microbiota. We hope that this review will be helpful for providing new ideas for the clinical application of TCMs in the treatment of AD.
Collapse
Affiliation(s)
- Jinyao Long
- Department of NeurologyXuanwu Hospital, Capital Medical UniversityBeijingChina
| | - Jiani Zhang
- School of Life SciencesBeijing University of Chinese MedicineBeijingChina
| | - Xin Zeng
- Department of NeurologyXuanwu Hospital, Capital Medical UniversityBeijingChina
| | - Min Wang
- Dongfang Hospital Beijing University of Chinese MedicineBeijingChina
| | - Ningqun Wang
- Department of NeurologyXuanwu Hospital, Capital Medical UniversityBeijingChina
| |
Collapse
|
15
|
Saadh MJ, Mustafa AN, Mustafa MA, S RJ, Dabis HK, Prasad GVS, Mohammad IJ, Adnan A, Idan AH. The role of gut-derived short-chain fatty acids in Parkinson's disease. Neurogenetics 2024; 25:307-336. [PMID: 39266892 DOI: 10.1007/s10048-024-00779-3] [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: 06/21/2024] [Accepted: 08/29/2024] [Indexed: 09/14/2024]
Abstract
The emerging function of short-chain fatty acids (SCFAs) in Parkinson's disease (PD) has been investigated in this article. SCFAs, which are generated via the fermentation of dietary fiber by gut microbiota, have been associated with dysfunction of the gut-brain axis and, neuroinflammation. These processes are integral to the development of PD. This article examines the potential therapeutic implications of SCFAs in the management of PD, encompassing their capacity to modulate gastrointestinal permeability, neuroinflammation, and neuronal survival, by conducting an extensive literature review. As a whole, this article emphasizes the potential therapeutic utility of SCFAs as targets for the management and treatment of PD.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan.
| | | | - Mohammed Ahmed Mustafa
- School of Pharmacy-Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University, Gangoh, Uttar Pradesh, 247341, India
- Department of Pharmacy, Arka Jain University, Jamshedpur, Jharkhand, 831001, India
| | - Renuka Jyothi S
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | | | - G V Siva Prasad
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra, Pradesh-531162, India
| | - Imad Jassim Mohammad
- College of Health and Medical Technology, National University of Science and Technology, Dhi Qar, 64001, Iraq
| | - Ahmed Adnan
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | | |
Collapse
|
16
|
Wu Q, Li W, Kwok LY, Lv H, Sun J, Sun Z. Regional variation and adaptive evolution in Bifidobacterium pseudocatenulatum: Insights into genomic and functional diversity in human gut. Food Res Int 2024; 192:114840. [PMID: 39147525 DOI: 10.1016/j.foodres.2024.114840] [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: 03/19/2024] [Revised: 07/16/2024] [Accepted: 07/26/2024] [Indexed: 08/17/2024]
Abstract
Bifidobacterium pseudocatenulatum is a prevalent gut microbe in humans of all ages and plays a crucial role in host health. However, its adaptive evolutionary characteristics remain poorly understood. This study analyzed the genome of 247 B. pseudocatenulatum isolates from Chinese, Vietnamese, Japanese and other region populations using population genomics and functional genomics. Our findings revealed high genetic heterogeneity and regional clustering within B. pseudocatenulatum isolates. Significant differences were observed in genome characteristics, phylogeny, and functional genes. Specifically, Chinese and Vietnamese isolates exhibited a higher abundance of genes involved in the metabolism of plant-derived carbohydrates (GH13, GH43, and GH5 enzyme families), aligning with the predominantly vegetable-, wheat- and fruit-based diets of these populations. Additionally, we found widespread transmission of antibiotic resistance genes (tetO and tetW) through mobile genetic elements, such as genomic islands (GIs), resulting in substantial intra-regional differences. Our findings highlight distinct adaptive evolution in B. pseudocatenulatum driven by gene specialization, possibly in response to regional variations in diet and lifestyle. This study sheds light on bifidobacteria colonization mechanisms in the host gut. IMPORTANCE: Gut microbiota, as a key link in the gut-brain axis, helps to maintain the health of the organism, among which, Bifidobacterium pseudocatenulatum (B. pseudocatenulatum) is an important constituent member of the gut microbiota, which plays an important role in maintaining the balance of gut microbiota. The probiotic properties of B. pseudocatenulatum have been widely elaborated, and in order to excavate its evolutionary features at the genomic level, here we focused on the genetic background and evolutionary mechanism of the B. pseudocatenulatum genomes isolated from the intestinal tracts of different populations. Ultimately, based on the phylogenetic tree, we found that B. pseudocatenulatum has high genetic diversity and regional clustering phenomenon, in which plant-derived carbohydrate metabolism genes (GH13, GH43, GH5) showed significant regional differences, and this genetic differentiation drove the adaptive evolution, which likely shaped by diet and lifestyle.
Collapse
Affiliation(s)
- Qiong Wu
- Key Laboratory of Dairy Biotechnology and Engineering (IMAU), Ministry of Education, Inner Mongolia Agricultural University, Hohhot, PR China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, PR China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, PR China
| | - Weicheng Li
- Key Laboratory of Dairy Biotechnology and Engineering (IMAU), Ministry of Education, Inner Mongolia Agricultural University, Hohhot, PR China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, PR China; Collaborative Innovative Center for Lactic Acid Bacteria and Fermented Dairy Products, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, PR China
| | - Lai-Yu Kwok
- Key Laboratory of Dairy Biotechnology and Engineering (IMAU), Ministry of Education, Inner Mongolia Agricultural University, Hohhot, PR China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, PR China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, PR China
| | - Huimin Lv
- Key Laboratory of Dairy Biotechnology and Engineering (IMAU), Ministry of Education, Inner Mongolia Agricultural University, Hohhot, PR China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, PR China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, PR China
| | - Jiaqi Sun
- Key Laboratory of Dairy Biotechnology and Engineering (IMAU), Ministry of Education, Inner Mongolia Agricultural University, Hohhot, PR China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, PR China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, PR China
| | - Zhihong Sun
- Key Laboratory of Dairy Biotechnology and Engineering (IMAU), Ministry of Education, Inner Mongolia Agricultural University, Hohhot, PR China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, PR China; Collaborative Innovative Center for Lactic Acid Bacteria and Fermented Dairy Products, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, PR China.
| |
Collapse
|
17
|
De Bruyn F, James K, Cottenet G, Dominick M, Katja J. Combining Bifidobacterium longum subsp. infantis and human milk oligosaccharides synergistically increases short chain fatty acid production ex vivo. Commun Biol 2024; 7:943. [PMID: 39098939 PMCID: PMC11298527 DOI: 10.1038/s42003-024-06628-1] [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: 01/30/2024] [Accepted: 07/24/2024] [Indexed: 08/06/2024] Open
Abstract
To enhance health benefits, a probiotic can be co-administered with a metabolizable prebiotic forming a synergistic synbiotic. We assessed the synergies resulting from combining Bifidobacterium longum subsp. infantis LMG 11588 and an age-adapted blend of six human milk oligosaccharides (HMOs) in ex vivo colonic incubation bioreactors seeded with fecal background microbiota from infant and toddler donors. When HMOs were combined with B. infantis LMG 11588, they were rapidly and completely consumed. This resulted in increased short chain fatty acid (SCFA) production compared to the summed SCFA production from individual ingredients (synergy). Remarkably, HMOs were partially consumed for specific infant donors in the absence of B. infantis LMG 11588, yet all donors showed increased SCFA production upon B. infantis LMG 11588 supplementation. We found specific bacterial taxa associated with the differential response pattern to HMOs. Our study shows the importance of carefully selecting pre- and probiotic into a synergistic synbiotic that could benefit infants.
Collapse
Affiliation(s)
- Florac De Bruyn
- Nestlé Research and Development, Nestléstrasse 3, CH-3510, Konolfingen, Switzerland.
| | - Kieran James
- Nestlé Research and Development, Nestléstrasse 3, CH-3510, Konolfingen, Switzerland
| | - Geoffrey Cottenet
- Nestlé Institute of Food Safety and Analytical Science, Nestlé Research, Route du Jorat 57, CH-1000, Lausanne, Switzerland
| | - Maes Dominick
- Nestlé Research and Development, Nestléstrasse 3, CH-3510, Konolfingen, Switzerland
| | - Johnson Katja
- Nestlé Research and Development, Nestléstrasse 3, CH-3510, Konolfingen, Switzerland
| |
Collapse
|
18
|
Xiao M, Zhang C, Duan H, Narbad A, Zhao J, Chen W, Zhai Q, Yu L, Tian F. Cross-feeding of bifidobacteria promotes intestinal homeostasis: a lifelong perspective on the host health. NPJ Biofilms Microbiomes 2024; 10:47. [PMID: 38898089 PMCID: PMC11186840 DOI: 10.1038/s41522-024-00524-6] [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: 12/29/2023] [Accepted: 06/07/2024] [Indexed: 06/21/2024] Open
Abstract
Throughout the life span of a host, bifidobacteria have shown superior colonization and glycan abilities. Complex glycans, such as human milk oligosaccharides and plant glycans, that reach the colon are directly internalized by the transport system of bifidobacteria, cleaved into simple structures by extracellular glycosyl hydrolase, and transported to cells for fermentation. The glycan utilization of bifidobacteria introduces cross-feeding activities between bifidobacterial strains and other microbiota, which are influenced by host nutrition and regulate gut homeostasis. This review discusses bifidobacterial glycan utilization strategies, focusing on the cross-feeding involved in bifidobacteria and its potential health benefits. Furthermore, the impact of cross-feeding on the gut trophic niche of bifidobacteria and host health is also highlighted. This review provides novel insights into the interactions between microbe-microbe and host-microbe.
Collapse
Affiliation(s)
- Meifang Xiao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Chuan Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Hui Duan
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Arjan Narbad
- Quadram Institute Bioscience, Norwich Research Park Colney, Norwich, Norfolk, NR4 7UA, UK
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Leilei Yu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China.
| | - Fengwei Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, 214122, P. R. China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China.
| |
Collapse
|
19
|
TAKESHITA K, TAKEI H, TANAKA S, HISHIKI H, IIJIMA Y, OGATA H, FUJISHIRO K, TOMINAGA T, KONNO Y, IWASE Y, ENDO M, ISHIWADA N, OSONE Y, TAKEMURA R, HAMADA H, SHIMOJO N. Effect of multi-strain bifidobacteria supplementation on intestinal microbiota development in low birth weight neonates: a randomized controlled trial. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2024; 43:352-358. [PMID: 39364130 PMCID: PMC11444860 DOI: 10.12938/bmfh.2023-093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 05/21/2024] [Indexed: 10/05/2024]
Abstract
Single-strain Bifidobacterium species are commonly used as probiotics with low birth weight neonates. However, the effectiveness and safety of multi-strain Bifidobacterium supplementation are not well known. Thirty-six neonates weighing less than 2,000 g (558-1,943 g) at birth and admitted to a neonatal intensive care unit were randomly assigned to receive a single strain or triple strains of Bifidobacterium with lactulose enterally for 4 weeks from birth. The relative abundances of Staphylococcus and Bifidobacterium in the fecal microbiota at weeks 1, 2, and 4 were investigated. Based on the study results, no significant difference was detected between the two groups in the abundance of Staphylococcus; however, the triple-strain group had significantly high abundances of Bifidobacterium at weeks 2 and 4. The fecal microbiota in the triple-strain group had significantly lower alpha diversity (Bifidobacterium-enriching) after week 4 and was different from that in the single-strain group, which showed a higher abundance of Clostridium. No severe adverse events occurred in either group during the study period. Although no significant difference was detected between single- and multi-strain bifidobacteria supplementation in the colonization of Staphylococcus in the fecal microbiota of the neonates, multi-strain bifidobacteria supplementation contributed toward early enrichment of the microbiota with bifidobacteria and suppression of other pathogenic bacteria, such as Clostridium spp.
Collapse
Affiliation(s)
- Kenichi TAKESHITA
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
- Department of Pediatrics, Chiba Rosai Hospital, 2-16
Tatsumidaihigashi, Ichihara-shi, Chiba 290-0003, Japan
| | - Haruka TAKEI
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
| | - Saori TANAKA
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
| | - Haruka HISHIKI
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
| | - Yuta IIJIMA
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
- Perinatal Medical Center, Chiba University Hospital, 1-8-1
Inohana, Chuo-ku, Chiba-shi, Chiba 260-8677, Japan
| | - Hitoshi OGATA
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
- Perinatal Medical Center, Chiba University Hospital, 1-8-1
Inohana, Chuo-ku, Chiba-shi, Chiba 260-8677, Japan
| | - Kensuke FUJISHIRO
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
- Perinatal Medical Center, Chiba University Hospital, 1-8-1
Inohana, Chuo-ku, Chiba-shi, Chiba 260-8677, Japan
| | - Takahiro TOMINAGA
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
- Perinatal Medical Center, Chiba University Hospital, 1-8-1
Inohana, Chuo-ku, Chiba-shi, Chiba 260-8677, Japan
| | - Yuki KONNO
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
- Perinatal Medical Center, Chiba University Hospital, 1-8-1
Inohana, Chuo-ku, Chiba-shi, Chiba 260-8677, Japan
| | - Yukiko IWASE
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
- Perinatal Medical Center, Chiba University Hospital, 1-8-1
Inohana, Chuo-ku, Chiba-shi, Chiba 260-8677, Japan
| | - Mamiko ENDO
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
- Perinatal Medical Center, Chiba University Hospital, 1-8-1
Inohana, Chuo-ku, Chiba-shi, Chiba 260-8677, Japan
| | - Naruhiko ISHIWADA
- Department of Infectious Diseases, Medical Mycology Research
Center, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8673, Japan
| | - Yoshiteru OSONE
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
- Perinatal Medical Center, Chiba University Hospital, 1-8-1
Inohana, Chuo-ku, Chiba-shi, Chiba 260-8677, Japan
| | - Ryo TAKEMURA
- Clinical and Translational Research Center, Keio University
Hospital, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hiromichi HAMADA
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
| | - Naoki SHIMOJO
- Center for Preventive Medical Sciences, Chiba University,
1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
| |
Collapse
|
20
|
Zhang J, Zhang C, Yu L, Tian F, Chen W, Zhai Q. Analysis of the key genes of Lactobacillus reuteri strains involved in the protection against alcohol-induced intestinal barrier damage. Food Funct 2024; 15:6629-6641. [PMID: 38812427 DOI: 10.1039/d4fo01796j] [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: 05/31/2024]
Abstract
Gastrointestinal inflammation and intestinal barrier function have important effects on human health. Alcohol, an important foodborne hazard factor, damages the intestinal barrier, increasing the risk of disease. Lactobacillus reuteri strains have been reported to reduce gastrointestinal inflammation and strengthen the intestinal barrier. In this study, we selected three anti-inflammatory L. reuteri strains to evaluate their role in the protection of the intestinal barrier and their immunomodulatory activity in a mouse model of gradient alcohol intake. Among the three strains tested (FSCDJY33M3, FGSZY33L6, and FCQHCL8L6), L. reuteri FSCDJY33M3 was found to protect the intestinal barrier most effectively, possibly due to its ability to reduce the expression of interleukin (IL)-1β, IL-6, and tumor necrosis factor-alpha (TNF-α) and increase the expression of tight junction proteins (occludin, claudin-3). Genomic analysis suggested that the protective effects of L. reuteri FSCDJY33M3 may be related to functional genes and glycoside hydrolases associated with energy production and conversion, amino acid transport and metabolism, carbohydrate transport and metabolism, and DNA replication, recombination, and repair. These genes include COG2856, COG1804, COG2071, and COG1061, which encode adenine deaminase, acyl-CoA transferases, glutamine amidotransferase, RNA helicase, and glycoside hydrolases, including GH13_20, GH53, and GH70. Our results identified functional genes that may be related to protection against alcohol-induced intestinal barrier damage, which might be useful for screening lactic acid bacterial strains that can protect the intestinal barrier.
Collapse
Affiliation(s)
- Jiayi Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Chengcheng Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Leilei Yu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| |
Collapse
|
21
|
Hermes GDA, Rasmussen C, Wellejus A. Variation in the Conservation of Species-Specific Gene Sets for HMO Degradation and Its Effects on HMO Utilization in Bifidobacteria. Nutrients 2024; 16:1893. [PMID: 38931248 PMCID: PMC11206791 DOI: 10.3390/nu16121893] [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: 05/13/2024] [Revised: 06/06/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Human milk provides essential nutrients for infants but also consists of human milk oligosaccharides (HMOs), which are resistant to digestion by the infant. Bifidobacteria are among the first colonizers, providing various health benefits for the host. This is largely facilitated by their ability to efficiently metabolize HMOs in a species-specific way. Nevertheless, these abilities can vary significantly by strain, and our understanding of the mechanisms applied by different strains from the same species remains incomplete. Therefore, we assessed the effects of strain-level genomic variation in HMO utilization genes on growth on HMOs in 130 strains from 10 species of human associated bifidobacteria. Our findings highlight the extent of genetic diversity between strains of the same species and demonstrate the effects on species-specific HMO utilization, which in most species is largely retained through the conservation of a core set of genes or the presence of redundant pathways. These data will help to refine our understanding of the genetic factors that contribute to the persistence of individual strains and will provide a better mechanistic rationale for the development and optimization of new early-life microbiota-modulating products to improve infant health.
Collapse
Affiliation(s)
- Gerben D. A. Hermes
- Human Health Research, Human Health Biosolutions, Novonesis, Kogle Alle 6, 2970 Hoersholm, Denmark (A.W.)
| | | | | |
Collapse
|
22
|
Sugiura S, Ikeda M, Nakamura Y, Mishima R, Morishita M, Nakayama J. Impact of vegetables on the microbiota of the rice bran pickling bed Nukadoko. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2024; 43:359-366. [PMID: 39364126 PMCID: PMC11444858 DOI: 10.12938/bmfh.2023-104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 05/21/2024] [Indexed: 10/05/2024]
Abstract
Nukadoko, a fermented rice bran bed for pickling vegetables called nukazuke, has a complex microbiota. Within it, deep interactions between the microbiota of the pickled vegetables and nukadoko characterize and control the qualities of both products. To address this notion, we monitored the changes in the microbiota of nukadoko and nukazuke while pickling different vegetables. Raw or roasted rice bran was mixed with salted water and fermented at 24°C for 40 days, following which different species of vegetable, Cucumis sativus var. sativus, Brassica oleracea var. capitata, or Raphanus sativus var. hortensis, were pickled. The microbial composition of the washing solution of fresh vegetables, as well as that of the nukadoko and nukazuke for each vegetable, was analyzed by amplicon sequencing of 16S rRNA genes. Although the microbiota of nukadoko varied depending on the species of pickled vegetables, no transcolonization of any species of bacteria from fresh vegetables to nukadoko was observed. However, some lactic acid bacterium (LAB) species eventually dominated the microbiota of both nukazuke and matured nukadoko, although they were not detected in either the fresh vegetables or rice bran. Particularly, Lactiplantibacillus plantarum was dominant among all pairs of pickled vegetables and matured nukadoko, whereas the transcolonization of some other LAB species was observed in a pickled vegetable-specific manner. Staphylococcus xylosus was observed to some extent in each nukadoko, yet it was not detected in any nukazuke. Overall, a LAB-dominant microbiota was established in both nukadoko and nukazuke in an underlying process that was different but partly common among vegetables.
Collapse
Affiliation(s)
- Shunsaku Sugiura
- Research Institute of Pickles Function, Tokai Pickling Co., Ltd., 78-1 Mukaigo, Mukokusama, Toyohashi, Aichi 441-8142, Japan
| | - Mika Ikeda
- Research Institute of Pickles Function, Tokai Pickling Co., Ltd., 78-1 Mukaigo, Mukokusama, Toyohashi, Aichi 441-8142, Japan
| | - Yuichi Nakamura
- Research Institute of Pickles Function, Tokai Pickling Co., Ltd., 78-1 Mukaigo, Mukokusama, Toyohashi, Aichi 441-8142, Japan
| | - Riko Mishima
- Laboratory of Microbial Technology, Division of Applied Molecular Microbiology and Biomass Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Mika Morishita
- Research Institute of Pickles Function, Tokai Pickling Co., Ltd., 78-1 Mukaigo, Mukokusama, Toyohashi, Aichi 441-8142, Japan
| | - Jiro Nakayama
- Laboratory of Microbial Technology, Division of Applied Molecular Microbiology and Biomass Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| |
Collapse
|
23
|
Tanaka S, Takahashi M, Takeshita K, Nagasawa K, Takei H, Sato H, Hishiki H, Ishiwada N, Hamada H, Kadota Y, Tochio T, Ishida T, Sasaki K, Tomita M, Osone Y, Takemura R, Shimojo N. The prebiotic effect of 1-kestose in low-birth-weight neonates taking bifidobacteria: a pilot randomized trial in comparison with lactulose. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2024; 43:329-335. [PMID: 39364124 PMCID: PMC11444857 DOI: 10.12938/bmfh.2023-079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 04/29/2024] [Indexed: 10/05/2024]
Abstract
Probiotics such as bifidobacteria have been given to low-birth-weight neonates (LBWNs) at risk for a disrupted gut microbiota leading to the development of serious diseases such necrotizing enterocolitis. Recently prebiotics such as lactulose are used together with bifidobacteria as synbiotics. However, faster and more powerful bifidobacteria growth is desired for better LBWN outcomes. The prebiotic 1-kestose has a higher selective growth-promoting effect on bifidobacteria and lactic acid bacteria in vitro among several oligosaccharides. Twenty-six premature neonates (less than 2,000 g) admitted to a neonatal intensive care unit (NICU) were randomly assigned to receive Bifidobacterium breve M16-V with either 1-kestose or lactulose once a day for four weeks from birth. A 16S rRNA gene analysis revealed similar increases in alpha-diversity from 7 to 28 days in both groups. The most dominant genus on both days was Bifidobacterium in both groups, with no significant difference between the two groups. Quantitative PCR analysis revealed that the number of Staphylococcus aureus tended to be lower in the 1-kestose group than in the lactulose group at 28 days. The number of Escherichia coli was higher in the 1-kestose group at 7 days. The copy number of total bacteria in the 1-kestose group was significantly higher than that in the lactulose group at 3 time points, 7, 14, and 28 days. No severe adverse events occurred in either group during the study period. l-Ketose may offer an alternative option to lactulose as a prebiotic to promote the development of gut microbiota in LBWNs.
Collapse
Affiliation(s)
- Saori Tanaka
- Department of Pediatrics, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
- Department of Neonatology, Kimitsu Chuo Hospital, 1010 Sakurai, Kisarazu-shi, Chiba 292-8535, Japan
| | - Mayuko Takahashi
- B Food Science Co., Ltd., 24-12 Kitahama-machi, Chita-shi, Aichi 478-0046, Japan
| | - Kenichi Takeshita
- Department of Pediatrics, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
| | - Koo Nagasawa
- Department of Pediatrics, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
| | - Haruka Takei
- Department of Pediatrics, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
| | - Hironori Sato
- Department of Pediatrics, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
| | - Haruka Hishiki
- Department of Pediatrics, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
| | - Naruhiko Ishiwada
- Department of Infectious Diseases, Medical Mycology Research Center, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8673, Japan
| | - Hiromichi Hamada
- Department of Pediatrics, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
| | - Yoshihiro Kadota
- B Food Science Co., Ltd., 24-12 Kitahama-machi, Chita-shi, Aichi 478-0046, Japan
| | - Takumi Tochio
- B Food Science Co., Ltd., 24-12 Kitahama-machi, Chita-shi, Aichi 478-0046, Japan
| | - Tomoki Ishida
- Department of Neonatology, Kimitsu Chuo Hospital, 1010 Sakurai, Kisarazu-shi, Chiba 292-8535, Japan
| | - Koh Sasaki
- Department of Neonatology, Kimitsu Chuo Hospital, 1010 Sakurai, Kisarazu-shi, Chiba 292-8535, Japan
| | - Mika Tomita
- Department of Neonatology, Kimitsu Chuo Hospital, 1010 Sakurai, Kisarazu-shi, Chiba 292-8535, Japan
| | - Yoshiteru Osone
- Department of Pediatrics, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
- Perinatal Medical Center, Chiba University Hospital, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8677, Japan
| | - Ryo Takemura
- Clinical and Translational Research Center, Keio University Hospital, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Naoki Shimojo
- Center for Preventive Medical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
| |
Collapse
|
24
|
Perdijk O, Azzoni R, Marsland BJ. The microbiome: an integral player in immune homeostasis and inflammation in the respiratory tract. Physiol Rev 2024; 104:835-879. [PMID: 38059886 DOI: 10.1152/physrev.00020.2023] [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: 05/02/2023] [Revised: 11/07/2023] [Accepted: 11/30/2023] [Indexed: 12/08/2023] Open
Abstract
The last decade of microbiome research has highlighted its fundamental role in systemic immune and metabolic homeostasis. The microbiome plays a prominent role during gestation and into early life, when maternal lifestyle factors shape immune development of the newborn. Breast milk further shapes gut colonization, supporting the development of tolerance to commensal bacteria and harmless antigens while preventing outgrowth of pathogens. Environmental microbial and lifestyle factors that disrupt this process can dysregulate immune homeostasis, predisposing infants to atopic disease and childhood asthma. In health, the low-biomass lung microbiome, together with inhaled environmental microbial constituents, establishes the immunological set point that is necessary to maintain pulmonary immune defense. However, in disease perturbations to immunological and physiological processes allow the upper respiratory tract to act as a reservoir of pathogenic bacteria, which can colonize the diseased lung and cause severe inflammation. Studying these host-microbe interactions in respiratory diseases holds great promise to stratify patients for suitable treatment regimens and biomarker discovery to predict disease progression. Preclinical studies show that commensal gut microbes are in a constant flux of cell division and death, releasing microbial constituents, metabolic by-products, and vesicles that shape the immune system and can protect against respiratory diseases. The next major advances may come from testing and utilizing these microbial factors for clinical benefit and exploiting the predictive power of the microbiome by employing multiomics analysis approaches.
Collapse
Affiliation(s)
- Olaf Perdijk
- Department of Immunology, School of Translational Science, Monash University, Melbourne, Victoria, Australia
| | - Rossana Azzoni
- Department of Immunology, School of Translational Science, Monash University, Melbourne, Victoria, Australia
| | - Benjamin J Marsland
- Department of Immunology, School of Translational Science, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
25
|
Lordan C, Roche AK, Delsing D, Nauta A, Groeneveld A, MacSharry J, Cotter PD, van Sinderen D. Linking human milk oligosaccharide metabolism and early life gut microbiota: bifidobacteria and beyond. Microbiol Mol Biol Rev 2024; 88:e0009423. [PMID: 38206006 PMCID: PMC10966949 DOI: 10.1128/mmbr.00094-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024] Open
Abstract
SUMMARYHuman milk oligosaccharides (HMOs) are complex, multi-functional glycans present in human breast milk. They represent an intricate mix of heterogeneous structures which reach the infant intestine in an intact form as they resist gastrointestinal digestion. Therefore, they confer a multitude of benefits, directly and/or indirectly, to the developing neonate. Certain bifidobacterial species, being among the earliest gut colonizers of breast-fed infants, have an adapted functional capacity to metabolize various HMO structures. This ability is typically observed in infant-associated bifidobacteria, as opposed to bifidobacteria associated with a mature microbiota. In recent years, information has been gleaned regarding how these infant-associated bifidobacteria as well as certain other taxa are able to assimilate HMOs, including the mechanistic strategies enabling their acquisition and consumption. Additionally, complex metabolic interactions occur between microbes facilitated by HMOs, including the utilization of breakdown products released from HMO degradation. Interest in HMO-mediated changes in microbial composition and function has been the focal point of numerous studies, in recent times fueled by the availability of individual biosynthetic HMOs, some of which are now commonly included in infant formula. In this review, we outline the main HMO assimilatory and catabolic strategies employed by infant-associated bifidobacteria, discuss other taxa that exhibit breast milk glycan degradation capacity, and cover HMO-supported cross-feeding interactions and related metabolites that have been described thus far.
Collapse
Affiliation(s)
- Cathy Lordan
- Teagasc Food Research Centre, Fermoy, Co Cork, Ireland
| | - Aoife K. Roche
- APC Microbiome Ireland, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | | | - Arjen Nauta
- FrieslandCampina, Amersfoort, the Netherlands
| | | | - John MacSharry
- APC Microbiome Ireland, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Paul D. Cotter
- Teagasc Food Research Centre, Fermoy, Co Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | - Douwe van Sinderen
- APC Microbiome Ireland, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| |
Collapse
|
26
|
van de Wouw M, Wang Y, Workentine ML, Vaghef-Mehrabani E, Barth D, Mercer EM, Dewey D, Arrieta MC, Reimer RA, Tomfohr-Madsen L, Giesbrecht GF. Cluster-specific associations between the gut microbiota and behavioral outcomes in preschool-aged children. MICROBIOME 2024; 12:60. [PMID: 38515179 PMCID: PMC10956200 DOI: 10.1186/s40168-024-01773-5] [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: 04/28/2023] [Accepted: 01/31/2024] [Indexed: 03/23/2024]
Abstract
BACKGROUND The gut microbiota is recognized as a regulator of brain development and behavioral outcomes during childhood. Nonetheless, associations between the gut microbiota and behavior are often inconsistent among studies in humans, perhaps because many host-microbe relationships vary widely between individuals. This study aims to stratify children based on their gut microbiota composition (i.e., clusters) and to identify novel gut microbiome cluster-specific associations between the stool metabolomic pathways and child behavioral outcomes. METHODS Stool samples were collected from a community sample of 248 typically developing children (3-5 years). The gut microbiota was analyzed using 16S sequencing while LC-MS/MS was used for untargeted metabolomics. Parent-reported behavioral outcomes (i.e., Adaptive Skills, Internalizing, Externalizing, Behavioral Symptoms, Developmental Social Disorders) were assessed using the Behavior Assessment System for Children (BASC-2). Children were grouped based on their gut microbiota composition using the Dirichlet multinomial method, after which differences in the metabolome and behavioral outcomes were investigated. RESULTS Four different gut microbiota clusters were identified, where the cluster enriched in both Bacteroides and Bifidobacterium (Ba2) had the most distinct stool metabolome. The cluster characterized by high Bifidobacterium abundance (Bif), as well as cluster Ba2, were associated with lower Adaptive Skill scores and its subcomponent Social Skills. Cluster Ba2 also had significantly lower stool histidine to urocanate turnover, which in turn was associated with lower Social Skill scores in a cluster-dependent manner. Finally, cluster Ba2 had increased levels of compounds involved in Galactose metabolism (i.e., stachyose, raffinose, alpha-D-glucose), where alpha-D-glucose was associated with the Adaptive Skill subcomponent Daily Living scores (i.e., ability to perform basic everyday tasks) in a cluster-dependent manner. CONCLUSIONS These data show novel associations between the gut microbiota, its metabolites, and behavioral outcomes in typically developing preschool-aged children. Our results support the concept that cluster-based groupings could be used to develop more personalized interventions to support child behavioral outcomes. Video Abstract.
Collapse
Affiliation(s)
- Marcel van de Wouw
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
| | - Yanan Wang
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
- Microbiomes for One Systems Health, Health & Biosecurity, CSIRO, Adelaide, SA, Australia
| | - Matthew L Workentine
- Faculty of Veterinary Medicine, UCVM Bioinformatics, University of Calgary, Calgary, Alberta, Canada
| | - Elnaz Vaghef-Mehrabani
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute (ACHRI), University of Calgary, Calgary, Alberta, Canada
| | - Delaney Barth
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
| | - Emily M Mercer
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, University of Calgary, Calgary, Alberta, Canada
| | - Deborah Dewey
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute (ACHRI), University of Calgary, Calgary, Alberta, Canada
- Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, Alberta, Canada
| | - Marie-Claire Arrieta
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, University of Calgary, Calgary, Alberta, Canada
| | - Raylene A Reimer
- Alberta Children's Hospital Research Institute (ACHRI), University of Calgary, Calgary, Alberta, Canada
- Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Lianne Tomfohr-Madsen
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute (ACHRI), University of Calgary, Calgary, Alberta, Canada
- Faculty of Education, University of British Columbia, Vancouver, British Columbia, Canada
| | - Gerald F Giesbrecht
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada.
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada.
- Alberta Children's Hospital Research Institute (ACHRI), University of Calgary, Calgary, Alberta, Canada.
- Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
27
|
Matsuzaki C, Takagi H, Saiga S, Kinoshita Y, Yamaguchi M, Higashimura Y, Yamamoto K, Yamaguchi M. Prebiotic effect of galacto- N-biose on the intestinal lactic acid bacteria as enhancer of acetate production and hypothetical colonization. Appl Environ Microbiol 2024; 90:e0144523. [PMID: 38411084 PMCID: PMC10952502 DOI: 10.1128/aem.01445-23] [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: 08/22/2023] [Accepted: 01/28/2024] [Indexed: 02/28/2024] Open
Abstract
Galacto-N-biose (GNB) is an important core structure of glycan of mucin glycoproteins in the gastrointestinal (GI) mucosa. Because certain beneficial bacteria inhabiting the GI tract, such as bifidobacteria and lactic acid bacteria, harbor highly specialized GNB metabolic capabilities, GNB is considered a promising prebiotic for nourishing and manipulating beneficial bacteria in the GI tract. However, the precise interactions between GNB and beneficial bacteria and their accompanying health-promoting effects remain elusive. First, we evaluated the proliferative tendency of beneficial bacteria and their production of beneficial metabolites using gut bacterial strains. By comparing the use of GNB, glucose, and inulin as carbon sources, we found that GNB enhanced acetate production in Lacticaseibacillus casei, Lacticaseibacillus rhamnosus, Lactobacillus gasseri, and Lactobacillus johnsonii. The ability of GNB to promote acetate production was also confirmed by RNA-seq analysis, which indicated the upregulation of gene clusters that catalyze the deacetylation of N-acetylgalactosamine-6P and biosynthesize acetyl-CoA from pyruvate, both of which result in acetate production. To explore the in vivo effect of GNB in promoting acetate production, antibiotic-treated BALB/cA mice were administered with GNB with L. rhamnosus, resulting in a fecal acetate content that was 2.7-fold higher than that in mice administered with only L. rhamnosus. Moreover, 2 days after the last administration, a 3.7-fold higher amount of L. rhamnosus was detected in feces administered with GNB with L. rhamnosus than in feces administered with only L. rhamnosus. These findings strongly suggest the prebiotic potential of GNB in enhancing L. rhamnosus colonization and converting L. rhamnosus into higher acetate producers in the GI tract. IMPORTANCE Specific members of lactic acid bacteria, which are commonly used as probiotics, possess therapeutic properties that are vital for human health enhancement by producing immunomodulatory metabolites such as exopolysaccharides, short-chain fatty acids, and bacteriocins. The long residence time of probiotic lactic acid bacteria in the GI tract prolongs their beneficial health effects. Moreover, the colonization property is also desirable for the application of probiotics in mucosal vaccination to provoke a local immune response. In this study, we found that GNB could enhance the beneficial properties of intestinal lactic acid bacteria that inhabit the human GI tract, stimulating acetate production and promoting intestinal colonization. Our findings provide a rationale for the addition of GNB to lactic acid bacteria-based functional foods. This has also led to the development of therapeutics supported by more rational prebiotic and probiotic selection, leading to an improved healthy lifestyle for humans.
Collapse
Affiliation(s)
- Chiaki Matsuzaki
- Research Institute for Bioresources and Biotechnology, Ishikawa Prefectural University, Nonoichi, Ishikawa, Japan
| | - Hiroki Takagi
- Department of Production Science, Ishikawa Prefectural University, Nonoichi, Ishikawa, Japan
| | - Sorachi Saiga
- Department of Production Science, Ishikawa Prefectural University, Nonoichi, Ishikawa, Japan
| | - Yuun Kinoshita
- Research Institute for Bioresources and Biotechnology, Ishikawa Prefectural University, Nonoichi, Ishikawa, Japan
| | - Misako Yamaguchi
- Department of Organic Bio Chemistry, Faculty of Education, Wakayama University, Wakayama, Japan
| | - Yasuki Higashimura
- Department of Food Science, Ishikawa Prefectural University, Nonoichi, Ishikawa, Japan
| | - Kenji Yamamoto
- Center for Innovative and Joint Research, Wakayama University, Wakayama, Japan
| | - Masanori Yamaguchi
- Department of Organic Bio Chemistry, Faculty of Education, Wakayama University, Wakayama, Japan
| |
Collapse
|
28
|
Kuntz S, Kunz C, Borsch C, Hill D, Morrin S, Buck R, Rudloff S. Influence of microbially fermented 2´-fucosyllactose on neuronal-like cell activity in an in vitro co-culture system. Front Nutr 2024; 11:1351433. [PMID: 38389793 PMCID: PMC10881714 DOI: 10.3389/fnut.2024.1351433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 01/18/2024] [Indexed: 02/24/2024] Open
Abstract
Scope 2´-Fucosyllactose (2´-FL), the most abundant oligosaccharide in human milk, plays an important role in numerous biological functions, including improved learning. It is not clear, however, whether 2´-FL or a cleavage product could influence neuronal cell activity. Thus, we investigated the effects of 2´-FL, its monosaccharide fucose (Fuc), and microbial fermented 2´-FL and Fuc on the parameters of neuronal cell activity in an intestinal-neuronal transwell co-culture system in vitro. Methods Native 13C-labeled 2´-FL and 13C-Fuc or their metabolites, fermented with Bifidobacterium (B.) longum ssp. infantis and B. breve, which were taken from the lag-, log- and stationary (stat-) growth phases of batch cultures, were applied to the apical compartment of the co-culture system with Caco-2 cells representing the intestinal layer and all-trans-retinoic acid-differentiated SH-SY5Y (SH-SY5YATRA) cells mimicking neuronal-like cells. After 3 h of incubation, the culture medium in the basal compartment was monitored for 13C enrichment by using elemental analysis isotope-ratio mass spectrometry (EA-IRMS) and effects on cell viability, plasma, and mitochondrial membrane potential. The neurotransmitter activation (BDNF, GABA, choline, and glutamate) of SH-SY5YATRA cells was also determined. Furthermore, these effects were also measured by the direct application of 13C-2´-FL and 13C-Fuc to SH-SY5YATRA cells. Results While no effects on neuronal-like cell activities were observed after intact 2´-FL or Fuc was incubated with SH-SY5YATRA cells, supernatants from the stat-growth phase of 2´-FL, fermented by B. longum ssp. infantis alone and together with B. breve, significantly induced BDNF release from SH-SY5YATRA cells. No such effects were found for 2´-FL, Fuc, or their fermentation products from B. breve. The BDNF release occurred from an enhanced vesicular release, which was confirmed by the use of the Ca2+-channel blocker verapamil. Concomitant with this event, 13C enrichment was also observed in the basal compartment when supernatants from the stat-growth phase of fermentation by B. longum ssp. infantis alone or together with B. breve were used. Conclusion The results obtained in this study suggest that microbial products of 2´-FL rather than the oligosaccharide itself may influence neuronal cell activities.
Collapse
Affiliation(s)
- Sabine Kuntz
- Department of Nutritional Science, Justus Liebig University Giessen, Giessen, Germany
| | - Clemens Kunz
- Department of Nutritional Science, Justus Liebig University Giessen, Giessen, Germany
| | - Christian Borsch
- Department of Nutritional Science, Justus Liebig University Giessen, Giessen, Germany
| | - David Hill
- Abbott, Nutrition Division, Columbus, OH, United States
| | - Sinéad Morrin
- Abbott, Nutrition Division, Columbus, OH, United States
| | - Rachael Buck
- Abbott, Nutrition Division, Columbus, OH, United States
| | - Silvia Rudloff
- Department of Nutritional Science, Justus Liebig University Giessen, Giessen, Germany
- Department of Pediatrics, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
29
|
Peled S, Freilich S, Hanani H, Kashi Y, Livney YD. Next-generation prebiotics: Maillard-conjugates of 2'-fucosyllactose and lactoferrin hydrolysates beneficially modulate gut microbiome composition and health promoting activity in a murine model. Food Res Int 2024; 177:113830. [PMID: 38225111 DOI: 10.1016/j.foodres.2023.113830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/01/2023] [Accepted: 12/05/2023] [Indexed: 01/17/2024]
Abstract
Current prebiotics are predominantly carbohydrates. However, great competition exists among gut microbes for the scarce protein in the colon, as most consumed protein is digested and absorbed in the small intestine. Herein we evaluated in-vivo novel next-generation prebiotics: protein-containing-prebiotics, for selectively-targeted delivery of protein to colonic probiotics, to boost their growth. This system is based on micellar-particles, composed of Maillard-glycoconjugates of 2'-Fucosyllactose (2'-FL, human-milk-oligosaccharide) shell, engulfing lactoferrin peptic-then-tryptic hydrolysate (LFH) core. This core-shell structure lowers protein-core digestibility, while the prebiotic glycans are hypothesized to serve as molecular-recognition ligands for selectively targeting probiotics. To study the efficacy of this novel prebiotic, we fed C57BL/6JRccHsd mice with either 2'-FL-LFH Maillard-glycoconjugates, unconjugated components (control), or saline (blank). Administration of 2'-FL-LFH significantly increased the levels of short-chain-fatty-acids (SCFAs)-producing bacterial families (Ruminococcaceae, Lachnospiraceae) and genus (Odoribacter) and the production of the health-related metabolites, SCFAs, compared to the unconjugated components and to saline. The SCFAs-producing genus Prevotella significantly increased upon 2'-FL-LFH consumption, compared to only moderate increase in the unconjugated components. Interestingly, the plasma-levels of inflammation-inducing lipopolysaccharides (LPS), which indicate increased gut-permeability, were significantly lower in the 2'-FL-LFH group compared to the unconjugated-components and the saline groups. We found that Maillard-glycoconjugates of 2'-FL-LFH can serve as novel protein-containing prebiotics, beneficially modulating gut microbial composition and its metabolic activity, thereby contributing to host health more effectively than the conventional carbohydrate-only prebiotics.
Collapse
Affiliation(s)
- Stav Peled
- Laboratory of Biopolymers for Food and Health, Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Shay Freilich
- Laboratory of Applied Genomics, Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Hila Hanani
- Laboratory of Applied Genomics, Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Yechezkel Kashi
- Laboratory of Applied Genomics, Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Yoav D Livney
- Laboratory of Biopolymers for Food and Health, Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel.
| |
Collapse
|
30
|
Catassi G, Aloi M, Giorgio V, Gasbarrini A, Cammarota G, Ianiro G. The Role of Diet and Nutritional Interventions for the Infant Gut Microbiome. Nutrients 2024; 16:400. [PMID: 38337684 PMCID: PMC10857663 DOI: 10.3390/nu16030400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/14/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
The infant gut microbiome plays a key role in the healthy development of the human organism and appears to be influenced by dietary practices through multiple pathways. First, maternal diet during pregnancy and infant nutrition significantly influence the infant gut microbiota. Moreover, breastfeeding fosters the proliferation of beneficial bacteria, while formula feeding increases microbial diversity. The timing of introducing solid foods also influences gut microbiota composition. In preterm infants the gut microbiota development is influenced by multiple factors, including the time since birth and the intake of breast milk, and interventions such as probiotics and prebiotics supplementation show promising results in reducing morbidity and mortality in this population. These findings underscore the need for future research to understand the long-term health impacts of these interventions and for further strategies to enrich the gut microbiome of formula-fed and preterm infants.
Collapse
Affiliation(s)
- Giulia Catassi
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (G.C.); (A.G.); (G.C.)
- Pediatric Gastroenterology and Liver Unit, Sapienza University of Rome, Umberto I Hospital, 00161 Rome, Italy;
| | - Marina Aloi
- Pediatric Gastroenterology and Liver Unit, Sapienza University of Rome, Umberto I Hospital, 00161 Rome, Italy;
| | - Valentina Giorgio
- Department of Woman and Child Health and Public Health, UOC Pediatria, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy;
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (G.C.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Giovanni Cammarota
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (G.C.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Gianluca Ianiro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (G.C.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|
31
|
Sato Y, Kanayama M, Nakajima S, Hishida Y, Watanabe Y. Sialyllactose Enhances the Short-Chain Fatty Acid Production and Barrier Function of Gut Epithelial Cells via Nonbifidogenic Modification of the Fecal Microbiome in Human Adults. Microorganisms 2024; 12:252. [PMID: 38399656 PMCID: PMC10892346 DOI: 10.3390/microorganisms12020252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/12/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
Although various benefits of human milk oligosaccharides (HMOs) have been reported, such as promoting Bifidobacterium growth in the infant gut, their effects on adults have not been fully studied. This study investigated the effects of two types of sialyllactose, 3'-sialyllactose (3'-SL) and 6'-sialyllactose (6'-SL), on the adult intestinal microbiome using the simulator of human intestinal microbial ecosystem (SHIME®), which can simulate human gastrointestinal conditions. HPLC metabolite analysis showed that sialyllactose (SL) supplementation increased the short-chain fatty acid content of SHIME culture broth. Moreover, 16S rRNA gene sequencing analysis revealed that SL promoted the growth of Phascolarctobacterium and Lachnospiraceae, short-chain fatty acid-producing bacteria, but not the growth of Bifidobacterium. Altogether, both types of SL stimulated an increase in short-chain fatty acids, including propionate and butyrate. Additionally, SHIME culture supernatant supplemented with SL improved the intestinal barrier function in Caco-2 cell monolayers. These results suggest that SL could act as a unique prebiotic among other HMOs with a nonbifidogenic effect, resulting in intestinal barrier protection.
Collapse
Affiliation(s)
- Yohei Sato
- Institute of Health Science, Kirin Holdings Co., Ltd., 2-26-1 Muraoka-Higashi, Fujisawa 251-8555, Japan; (Y.S.); (M.K.); (Y.H.)
| | - Masaya Kanayama
- Institute of Health Science, Kirin Holdings Co., Ltd., 2-26-1 Muraoka-Higashi, Fujisawa 251-8555, Japan; (Y.S.); (M.K.); (Y.H.)
| | - Shiori Nakajima
- Health Science Business Department, Kirin Holdings Co., Ltd., 4-10-2 Nakano, Tokyo 164-0001, Japan;
| | - Yukihiro Hishida
- Institute of Health Science, Kirin Holdings Co., Ltd., 2-26-1 Muraoka-Higashi, Fujisawa 251-8555, Japan; (Y.S.); (M.K.); (Y.H.)
| | - Yuta Watanabe
- Institute of Health Science, Kirin Holdings Co., Ltd., 2-26-1 Muraoka-Higashi, Fujisawa 251-8555, Japan; (Y.S.); (M.K.); (Y.H.)
| |
Collapse
|
32
|
Lee YG, Jo HY, Lee DH, Yoon JW, Song YH, Kweon DH, Kim KH, Park YC, Seo JH. De novo biosynthesis of 2'-fucosyllactose by bioengineered Corynebacterium glutamicum. Biotechnol J 2024; 19:e2300461. [PMID: 37968827 DOI: 10.1002/biot.202300461] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/27/2023] [Accepted: 11/12/2023] [Indexed: 11/17/2023]
Abstract
2'-Fucosyllactose (2'-FL) which is well-known human milk oligosaccharide was biotechnologically synthesized using engineered Corynebacterium glutamicum, a GRAS microbial workhorse. By construction of the complete de novo pathway for GDP-L-fucose supply and heterologous expression of Escherichia coli lactose permease and Helicobacter pylori α-1,2-fucosyltransferase, bioengineered C. glutamicum BCGW_TL successfully biosynthesized 0.25 g L-1 2'-FL from glucose. The additional genetic perturbations including the expression of a putative 2'-FL exporter and disruption of the chromosomal pfkA gene allowed C. glutamicum BCGW_cTTLEΔP to produce 2.5 g L-1 2'-FL batchwise. Finally, optimized fed-batch cultivation of the BCGW_cTTLEΔP using glucose, fructose, and lactose resulted in 21.5 g L-1 2'-FL production with a productivity of 0.12 g L-1 •h, which were more than 3.3 times higher value relative to the batch culture of the BCGW_TL. Conclusively, it would be a groundwork to adopt C. glutamicum for biotechnological production of other food additives including human milk oligosaccharides.
Collapse
Affiliation(s)
- Ye-Gi Lee
- Department of Bio and Fermentation Convergence Technology and Center for Bioconvergence, Kookmin University, Seoul, South Korea
- Department of Agricultural Biotechnology and Center for Food Bioconvergence, Seoul National University, Seoul, South Korea
| | - Hae-Yong Jo
- Department of Agricultural Biotechnology and Center for Food Bioconvergence, Seoul National University, Seoul, South Korea
| | - Do-Haeng Lee
- Department of Agricultural Biotechnology and Center for Food Bioconvergence, Seoul National University, Seoul, South Korea
| | - Jong-Won Yoon
- Advanced Protein Technologies Corp. Yeongtong-gu, Suwon, Gyeonggi, South Korea
| | - Young-Ha Song
- Advanced Protein Technologies Corp. Yeongtong-gu, Suwon, Gyeonggi, South Korea
| | - Dae-Hyuk Kweon
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, South Korea
| | - Kyoung Heon Kim
- Department of Biotechnology, Graduate School, Korea University, Seoul, South Korea
| | - Yong-Cheol Park
- Department of Bio and Fermentation Convergence Technology and Center for Bioconvergence, Kookmin University, Seoul, South Korea
| | - Jin-Ho Seo
- Department of Agricultural Biotechnology and Center for Food Bioconvergence, Seoul National University, Seoul, South Korea
| |
Collapse
|
33
|
Al-Busaidi A, Alabri O, Alomairi J, ElSharaawy A, Al Lawati A, Al Lawati H, Das S. Gut Microbiota and Insulin Resistance: Understanding the Mechanism of Better Treatment of Type 2 Diabetes Mellitus. Curr Diabetes Rev 2024; 21:e170124225723. [PMID: 38243954 DOI: 10.2174/0115733998281910231231051814] [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: 09/25/2023] [Revised: 12/13/2023] [Accepted: 12/20/2023] [Indexed: 01/22/2024]
Abstract
Gut microbiota refers to the population of trillions of microorganisms present in the human intestine. The gut microbiota in the gastrointestinal system is important for an individual's good health and well-being. The possibility of an intrauterine colonization of the placenta further suggests that the fetal environment before birth may also affect early microbiome development. Various factors influence the gut microbiota. Dysbiosis of microbiota may be associated with various diseases. Insulin regulates blood glucose levels, and disruption of the insulin signaling pathway results in insulin resistance. Insulin resistance or hyperinsulinemia is a pathological state in which the insulin-responsive cells have a diminished response to the hormone compared to normal physiological responses, resulting in reduced glucose uptake by the tissue cells. Insulin resistance is an important cause of type 2 diabetes mellitus. While there are various factors responsible for the etiology of insulin resistance, dysbiosis of gut microbiota may be an important contributing cause for metabolic disturbances. We discuss the mechanisms in skeletal muscles, adipose tissue, liver, and intestine by which insulin resistance can occur due to gut microbiota's metabolites. A better understanding of gut microbiota may help in the effective treatment of type 2 diabetes mellitus and metabolic syndrome.
Collapse
Affiliation(s)
- Alsalt Al-Busaidi
- Department of Medicine, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland
| | - Omer Alabri
- Department of Medicine, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland
| | - Jaifar Alomairi
- Department of Medicine, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland
| | | | | | - Hanan Al Lawati
- Pharmacy Program, Department of Pharmaceutics, Oman College of Health Sciences, Muscat 113, Oman
| | - Srijit Das
- Department of Human & Clinical Anatomy, College of Medicine & Health Sciences, Sultan Qaboos University, Muscat, Oman
| |
Collapse
|
34
|
Zhang C, Yu L, Ma C, Jiang S, Zhang Y, Wang S, Tian F, Xue Y, Zhao J, Zhang H, Liu L, Chen W, Huang S, Zhang J, Zhai Q. A key genetic factor governing arabinan utilization in the gut microbiome alleviates constipation. Cell Host Microbe 2023; 31:1989-2006.e8. [PMID: 37992712 DOI: 10.1016/j.chom.2023.10.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 09/01/2023] [Accepted: 10/11/2023] [Indexed: 11/24/2023]
Abstract
Impaired gastrointestinal motility is associated with gut dysbiosis. Probiotics, such as Bifidobacteria, can improve this bowel disorder; however, efficacy is strain-dependent. We determine that a genetic factor, the abfA cluster governing arabinan utilization, in Bifidobacterium longum impacts treatment efficacy against functional constipation (FC). In mice with FC, B. longum, but not an abfA mutant, improved gastrointestinal transit time, an affect that was dependent upon dietary arabinan. abfA genes were identified in other commensal bacteria, whose effects in ameliorating murine FC were similarly abfA-dependent. In a double-blind, randomized, placebo-controlled clinical trial, supplementation with abfA-cluster-carrying B. longum, but not an abfA-deficient strain, enriched arabinan-utilization residents, increased beneficial metabolites, and improved FC symptoms. Across human cohorts, abfA-cluster abundance can predict FC, and transplantation of abfA cluster-enriched human microbiota to FC-induced germ-free mice improved gut motility. Collectively, these findings demonstrate a role for microbial abfA cluster in ameliorating FC, establishing principles for genomics-directed probiotic therapies.
Collapse
Affiliation(s)
- Chengcheng Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Leilei Yu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Chenchen Ma
- College of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou 570228, China
| | - Shuaiming Jiang
- College of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou 570228, China
| | - Yufeng Zhang
- Faculty of Dentistry, University of Hong Kong, Hong Kong SAR, China
| | - Shunhe Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yuzheng Xue
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Liming Liu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Shi Huang
- Faculty of Dentistry, University of Hong Kong, Hong Kong SAR, China.
| | - Jiachao Zhang
- College of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou 570228, China.
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
35
|
Lou YC, Rubin BE, Schoelmerich MC, DiMarco KS, Borges AL, Rovinsky R, Song L, Doudna JA, Banfield JF. Infant microbiome cultivation and metagenomic analysis reveal Bifidobacterium 2'-fucosyllactose utilization can be facilitated by coexisting species. Nat Commun 2023; 14:7417. [PMID: 37973815 PMCID: PMC10654741 DOI: 10.1038/s41467-023-43279-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/06/2023] [Indexed: 11/19/2023] Open
Abstract
The early-life gut microbiome development has long-term health impacts and can be influenced by factors such as infant diet. Human milk oligosaccharides (HMOs), an essential component of breast milk that can only be metabolized by some beneficial gut microorganisms, ensure proper gut microbiome establishment and infant development. However, how HMOs are metabolized by gut microbiomes is not fully elucidated. Isolate studies have revealed the genetic basis for HMO metabolism, but they exclude the possibility of HMO assimilation via synergistic interactions involving multiple organisms. Here, we investigate microbiome responses to 2'-fucosyllactose (2'FL), a prevalent HMO and a common infant formula additive, by establishing individualized microbiomes using fecal samples from three infants as the inocula. Bifidobacterium breve, a prominent member of infant microbiomes, typically cannot metabolize 2'FL. Using metagenomic data, we predict that extracellular fucosidases encoded by co-existing members such as Ruminococcus gnavus initiate 2'FL breakdown, thus critical for B. breve's growth. Using both targeted co-cultures and by supplementation of R. gnavus into one microbiome, we show that R. gnavus can promote extensive growth of B. breve through the release of lactose from 2'FL. Overall, microbiome cultivation combined with genome-resolved metagenomics demonstrates that HMO utilization can vary with an individual's microbiome.
Collapse
Affiliation(s)
- Yue Clare Lou
- Department of Plant and Microbial Biology, University of California, Berkeley, CA, USA
- Innovative Genomics Institute, University of California, Berkeley, CA, USA
| | - Benjamin E Rubin
- Innovative Genomics Institute, University of California, Berkeley, CA, USA
| | - Marie C Schoelmerich
- Innovative Genomics Institute, University of California, Berkeley, CA, USA
- Department of Environmental Systems Sciences, ETH Zurich, Zurich, Switzerland
| | - Kaden S DiMarco
- Innovative Genomics Institute, University of California, Berkeley, CA, USA
| | - Adair L Borges
- Innovative Genomics Institute, University of California, Berkeley, CA, USA
| | - Rachel Rovinsky
- Innovative Genomics Institute, University of California, Berkeley, CA, USA
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Leo Song
- Innovative Genomics Institute, University of California, Berkeley, CA, USA
| | - Jennifer A Doudna
- Innovative Genomics Institute, University of California, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Department of Chemistry, University of California, Berkeley, CA, USA
- Howard Hughes Medical Institute, University of California, Berkeley, CA, USA
| | - Jillian F Banfield
- Innovative Genomics Institute, University of California, Berkeley, CA, USA.
- Department of Earth and Planetary Science, University of California, Berkeley, CA, USA.
- Department of Environmental Science, Policy, and Management, University of California, Berkeley, CA, USA.
| |
Collapse
|
36
|
Yahagi K. Fucosylated human milk oligosaccharide-utilizing bifidobacteria regulate the gut organic acid profile of infants. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2023; 43:92-99. [PMID: 38562549 PMCID: PMC10981941 DOI: 10.12938/bmfh.2023-069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 10/11/2023] [Indexed: 04/04/2024]
Abstract
Bifidobacteria are the predominant bacteria in the infant gut and have beneficial effects on host physiology. Infant cohort studies have demonstrated that a higher abundance of bifidobacteria in the gut is associated with a reduced risk of disease. Recently, bifidobacteria-derived metabolites, such as organic acid, have been suggested to play crucial roles in host physiology. This review focuses on an investigation of longitudinal changes in the gut microbiota and organic acid concentrations over 2 years of life in 12 Japanese infants and aims to identify bifidobacteria that contribute to the production of organic acid in healthy infants. Acetate, lactate, and formate, which are rarely observed in adults, are characteristically observed during breast-fed infancy. Bifidobacterium longum subspecies infantis and the symbiosis of Bifidobacterium bifidum and Bifidobacterium breve efficiently produce these organic acids through metabolization of human milk oligosaccharide (HMO) with different strategies. These findings confirmed that HMO-utilizing bifidobacteria play an important role in regulating the gut organic acid profiles of infants.
Collapse
Affiliation(s)
- Kana Yahagi
- Yakult Central Institute, Yakult Honsha Co., Ltd., 5-11 Izumi, Kunitachi-shi, Tokyo 186-8650, Japan
| |
Collapse
|
37
|
Kiely LJ, Busca K, Lane JA, van Sinderen D, Hickey RM. Molecular strategies for the utilisation of human milk oligosaccharides by infant gut-associated bacteria. FEMS Microbiol Rev 2023; 47:fuad056. [PMID: 37793834 PMCID: PMC10629584 DOI: 10.1093/femsre/fuad056] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 09/14/2023] [Accepted: 10/03/2023] [Indexed: 10/06/2023] Open
Abstract
A number of bacterial species are found in high abundance in the faeces of healthy breast-fed infants, an occurrence that is understood to be, at least in part, due to the ability of these bacteria to metabolize human milk oligosaccharides (HMOs). HMOs are the third most abundant component of human milk after lactose and lipids, and represent complex sugars which possess unique structural diversity and are resistant to infant gastrointestinal digestion. Thus, these sugars reach the infant distal intestine intact, thereby serving as a fermentable substrate for specific intestinal microbes, including Firmicutes, Proteobacteria, and especially infant-associated Bifidobacterium spp. which help to shape the infant gut microbiome. Bacteria utilising HMOs are equipped with genes associated with their degradation and a number of carbohydrate-active enzymes known as glycoside hydrolase enzymes have been identified in the infant gut, which supports this hypothesis. The resulting degraded HMOs can also be used as growth substrates for other infant gut bacteria present in a microbe-microbe interaction known as 'cross-feeding'. This review describes the current knowledge on HMO metabolism by particular infant gut-associated bacteria, many of which are currently used as commercial probiotics, including the distinct strategies employed by individual species for HMO utilisation.
Collapse
Affiliation(s)
- Leonie Jane Kiely
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork P61C996, Ireland
- Health and Happiness Group, H&H Research, National Food Innovation Hub, Teagasc Moorepark, Fermoy, Co. Cork P61K202, Ireland
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12 YT20, Ireland
- School of Microbiology, University College Cork, Cork T12 YN60, Ireland
| | - Kizkitza Busca
- Health and Happiness Group, H&H Research, National Food Innovation Hub, Teagasc Moorepark, Fermoy, Co. Cork P61K202, Ireland
| | - Jonathan A Lane
- Health and Happiness Group, H&H Research, National Food Innovation Hub, Teagasc Moorepark, Fermoy, Co. Cork P61K202, Ireland
| | - Douwe van Sinderen
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12 YT20, Ireland
- School of Microbiology, University College Cork, Cork T12 YN60, Ireland
| | - Rita M Hickey
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork P61C996, Ireland
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12 YT20, Ireland
| |
Collapse
|
38
|
Li R, Roy R. Gut Microbiota and Its Role in Anti-aging Phenomenon: Evidence-Based Review. Appl Biochem Biotechnol 2023; 195:6809-6823. [PMID: 36930406 DOI: 10.1007/s12010-023-04423-y] [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] [Accepted: 03/07/2023] [Indexed: 03/18/2023]
Abstract
The gut microbiota widely varies from individual to individual, but the variation shows stability over a period of time. The presence of abundant bacterial taxa is a common structure that determines the microbiota of human being. The presence of this microbiota greatly varies from geographic location, sex, food habits and age. Microbiota existing within the gut plays a significant role in nutrient absorption, development of immunity, curing of diseases and various developmental phases. With change in age, chronology diversification and variation of gut microbiota are observed within human being. But it has been observed that with the enhancement of age the richness of the microbial diversity has shown a sharp decline. The enhancement of age also results in the drift of the characteristic of the microbes associated with the microbiota from commensals to pathogenic. Various studies have shown that age associated gut-dysbiosis may result in decrease in tlongevity along with unhealthy aging. The host signalling pathways regulate the presence of the gut microbiota and their longevity. The presence of various nutrients regulates the presence of various microbial species. Innate immunity can be triggered due to the mechanism of gut dysbiosis resulting in the development of various age-related pathological syndromes and early aging. The gut microbiota possesses the ability to communicate with the host system with the help of various types of biomolecules, epigenetic mechanisms and various types of signalling-independent pathways. Drift in this mechanism of communication may affect the life span along with the health of the host. Thus, this review would focus on the use of gut-microbiota in anti-aging and healthy conditions of the host system.
Collapse
Affiliation(s)
- Ruishan Li
- Guiyang Healthcare Vocational University, Guiyang, China.
| | - Rupak Roy
- SHRM Biotechnologies Pvt. Ltd, Kolkata, India
| |
Collapse
|
39
|
Zhu L, Li H, Luo T, Deng Z, Li J, Zheng L, Zhang B. Human Milk Oligosaccharides: A Critical Review on Structure, Preparation, Their Potential as a Food Bioactive Component, and Future Perspectives. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:15908-15925. [PMID: 37851533 DOI: 10.1021/acs.jafc.3c04412] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
Human milk is the gold standard for infant feeding. Human milk oligosaccharides (HMOs) are a unique group of oligosaccharides in human milk. Great interest in HMOs has grown in recent years due to their positive effects on various aspects of infant health. HMOs provide various physiologic functions, including establishing a balanced infant's gut microbiota, strengthening the gastrointestinal barrier, preventing infections, and potential support to the immune system. However, the clinical application of HMOs is challenging due to their specificity to human milk and the difficulties and high costs associated with their isolation and synthesis. Here, the differences in oligosaccharides in human and other mammalian milk are compared, and the synthetic strategies to access HMOs are summarized. Additionally, the potential use and molecular mechanisms of HMOs as a new food bioactive component in different diseases, such as infection, necrotizing enterocolitis, diabetes, and allergy, are critically reviewed. Finally, the current challenges and prospects of HMOs in basic research and application are discussed.
Collapse
Affiliation(s)
- Liuying Zhu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Hongyan Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Ting Luo
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Zeyuan Deng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Jing Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Liufeng Zheng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Bing Zhang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| |
Collapse
|
40
|
Sahin S, Buyuktiryaki M, Okur N, Akcan AB, Deveci MF, Yurttutan S, Gunes S, Anik A, Ozdemir R, Uygur O, Oncel MY. Effect of partially hydrolyzed synbiotic formula milk on weight gain of late preterm and term infants-a multicenter study. Front Pediatr 2023; 11:1270442. [PMID: 37928348 PMCID: PMC10623126 DOI: 10.3389/fped.2023.1270442] [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: 07/31/2023] [Accepted: 09/28/2023] [Indexed: 11/07/2023] Open
Abstract
Introduction Data on the effectiveness of hydrolyzed infant formula containing both pre- and probiotics (synbiotic formula) on the growth of infants is still scarce. This retrospective study was designed to evaluate the effect of a partially hydrolyzed synbiotic formula on growth parameters and the possible occurrence of major gastrointestinal adverse events or morbidities in infants born via cesarean section (C-section) delivery. Methods C-section-delivered term and late preterm infants who received either partially hydrolyzed synbiotic formula, standard formula, or maternal milk and followed at seven different hospitals from five different regions of Turkey, during a 1-year period with a minimum follow-up duration of 3 months were evaluated retrospectively. All the included infants were evaluated for their growth patterns and any kind of morbidity such as diarrhea, constipation, vomiting, infection, or history of hospitalization. Results A total of 198 infants (73 in the human milk group, 61 in the standard formula group, and 64 in the partially hydrolyzed synbiotic formula group) reached the final analysis. The groups were similar regarding their demographic and perinatal characteristics. No difference was observed between the three groups regarding gastrointestinal major side effects. Growth velocities of the infants in the human milk and partially hydrolyzed synbiotic formula groups during the first month of life were similar whereas the weight gain of infants in the standard formula group was significantly less than these two groups (p < 0.001). Growth velocities were similar among the three groups between 1st and 3rd months of age. Discussion A partially hydrolyzed synbiotic formula provided better weight gain in late-preterm and term infants who were delivered via C-section delivery compared to the standard formula during the first month of life. This weight gain was similar to the infants receiving exclusively human milk. This difference was not observed in length and head circumference gain. No difference was observed in any of the parameters during the 1st-3rd months of age. Specially formulated partially hydrolyzed synbiotic formulas may reverse at least some of the negative impacts of C-section delivery on the infant and help to provide better growth, especially during the early periods of life.
Collapse
Affiliation(s)
- Suzan Sahin
- Faculty of Medicine, Department of Pediatrics, Division of Neonatology, Izmir Democracy University, Izmir, Türkiye
| | - Mehmet Buyuktiryaki
- Faculty of Medicine, Department of Pediatrics, Division of Neonatology, Istanbul Medipol University, Istanbul, Türkiye
| | - Nilufer Okur
- Department of Pediatrics, Division of Neonatology, Gazi Yasargil Training and Research Hospital, Diyarbakir, Türkiye
| | - Abdullah Baris Akcan
- Faculty of Medicine, Department of Pediatrics, Division of Neonatology, Adnan Menderes University, Aydin, Türkiye
| | - Mehmet Fatih Deveci
- Faculty of Medicine, Turgut Ozal Medical Center, Department of Pediatrics, Division of Neonatology, Inonu University, Malatya, Türkiye
| | - Sadik Yurttutan
- Faculty of Medicine, Department of Pediatrics, Division of Neonatology, Kahramanmaras Sutcu Imam University, Kahramanmaras, Türkiye
| | - Sezgin Gunes
- Department of Pediatrics, Division of Neonatology, Buca Seyfi Demirsoy Training and Research Hospital, Izmir, Türkiye
| | - Ayse Anik
- Faculty of Medicine, Department of Pediatrics, Division of Neonatology, Adnan Menderes University, Aydin, Türkiye
| | - Ramazan Ozdemir
- Faculty of Medicine, Turgut Ozal Medical Center, Department of Pediatrics, Division of Neonatology, Inonu University, Malatya, Türkiye
| | - Ozgun Uygur
- Department of Pediatrics, Division of Neonatology, Tepecik Training and Research Hospital, Izmir Health Sciences University, Izmir, Türkiye
| | - Mehmet Yekta Oncel
- Faculty of Medicine, Department of Pediatrics, Division of Neonatology, Izmir Katip Celebi University, Izmir, Türkiye
| |
Collapse
|
41
|
Mills M, Lee S, Piperata BA, Garabed R, Choi B, Lee J. Household environment and animal fecal contamination are critical modifiers of the gut microbiome and resistome in young children from rural Nicaragua. MICROBIOME 2023; 11:207. [PMID: 37715296 PMCID: PMC10503196 DOI: 10.1186/s40168-023-01636-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 07/31/2023] [Indexed: 09/17/2023]
Abstract
BACKGROUND Early life plays a vital role in the development of the gut microbiome and subsequent health. While many factors that shape the gut microbiome have been described, including delivery mode, breastfeeding, and antibiotic use, the role of household environments is still unclear. Furthermore, the development of the gut antimicrobial resistome and its role in health and disease is not well characterized, particularly in settings with water insecurity and less sanitation infrastructure. RESULTS This study investigated the gut microbiome and resistome of infants and young children (ages 4 days-6 years) in rural Nicaragua using Oxford Nanopore Technology's MinION long-read sequencing. Differences in gut microbiome diversity and antibiotic resistance gene (ARG) abundance were examined for associations with host factors (age, sex, height for age z-score, weight for height z-score, delivery mode, breastfeeding habits) and household environmental factors (animals inside the home, coliforms in drinking water, enteric pathogens in household floors, fecal microbial source tracking markers in household floors). We identified anticipated associations of higher gut microbiome diversity with participant age and vaginal delivery. However, novel to this study were the significant, positive associations between ruminant and dog fecal contamination of household floors and gut microbiome diversity. We also identified greater abundance of potential pathogens in the gut microbiomes of participants with higher fecal contamination on their household floors. Path analysis revealed that water quality and household floor contamination independently and significantly influenced gut microbiome diversity when controlling for age. These gut microbiome contained diverse resistome, dominated by multidrug, tetracycline, macrolide/lincosamide/streptogramin, and beta-lactam resistance. We found that the abundance of ARGs in the gut decreased with age. The bacterial hosts of ARGs were mainly from the family Enterobacteriaceae, particularly Escherichia coli. CONCLUSIONS This study identified the role of household environmental contamination in the developing gut microbiome and resistome of young children and infants with a One Health perspective. We found significant relationships between host age, gut microbiome diversity, and the resistome. Understanding the impact of the household environment on the development of the resistome and microbiome in early life is essential to optimize the relationship between environmental exposure and human health. Video Abstract.
Collapse
Affiliation(s)
- Molly Mills
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University, Columbus, OH, USA
- Environmental Sciences Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Seungjun Lee
- Department of Food Science and Nutrition, College of Fisheries Science, Pukyong National University, Busan, Republic of Korea
| | - Barbara A Piperata
- Department of Anthropology, The Ohio State University, Columbus, OH, USA
| | - Rebecca Garabed
- Department of Veterinary Preventive Medicine, The Ohio State University, Columbus, OH, USA
| | - Boseung Choi
- Division of Big Data Science, Korea University, Sejong, Republic of Korea
| | - Jiyoung Lee
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University, Columbus, OH, USA.
- Environmental Sciences Graduate Program, The Ohio State University, Columbus, OH, USA.
- Department of Food Science & Technology, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
42
|
Liu S, Mao Y, Wang J, Tian F, Hill DR, Xiong X, Li X, Zhao Y, Wang S. Lactational and geographical variation in the concentration of six oligosaccharides in Chinese breast milk: a multicenter study over 13 months postpartum. Front Nutr 2023; 10:1267287. [PMID: 37731395 PMCID: PMC10508235 DOI: 10.3389/fnut.2023.1267287] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 08/23/2023] [Indexed: 09/22/2023] Open
Abstract
Introduction Understanding the variations of oligosaccharide in breast milk contribute to better study how human milk oligosaccharides (HMOs) play a role in health-promoting benefits in infants. Methods Six abundant HMOs, 2'-fucosyllactose (2'-FL), 3-fucosyllactose (3-FL), Lacto-N-tetraose (LNT), Lacto-N-neotetraose (LNnT), 3'-sialyllactose (3'-SL) and 6'-sialyllactose (6'-SL), in breast milk collected at 0-5 days, 10-15 days, 40-45 days, 200-240 days, and 300-400 days postpartum from six locations across China were analyzed using high-performance anion-exchange chromatography-pulsed amperometric detector. Results The concentration of individual HMO fluctuated dynamically during lactational stages. The median ranges of 2'-FL, 3-FL, LNT, LNnT, 3'-SL, and 6'-SL across the five lactational stages were 935-2865 mg/L, 206-1325 mg/L, 300-1473 mg/L, 32-317 mg/L, 106-228 mg/L, and 20-616 mg/L, respectively. The prominent variation was observed in the content of 6'-SL, which demonstrates a pattern of initial increase followed by a subsequent decrease. Among the five lactational stages, the transitional milk has the highest concentration, which was 31 times greater than the concentration in mature milk at 300-400 days postpartum, where the content is the lowest. Geographical location also influenced the content of HMOs. LNT and LNnT were the highest in mature milk of mothers from Lanzhou among the six sites at 40-240 days postpartum. Breast milks were categorized into two groups base on the abundance of 2'-FL (high and low). There was no significant difference in the proportions of high and low 2'-FL phenotypes among the six sites, and the percentages of high and low 2'-FL phenotypes were 79% and 21%, respectively, across all sites in China. Discussion This study provided a comprehensive dataset on 6 HMOs concentrations in Chinese breast milk during the extended postpartum period across a wide geographic range and stratified by high and low 2'-FL phenotypes.
Collapse
Affiliation(s)
- Shuang Liu
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin, China
| | - Yingyi Mao
- Abbott Nutrition Research & Development Center, Shanghai, China
| | - Jin Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin, China
| | - Fang Tian
- Abbott Nutrition Research & Development Center, Shanghai, China
| | - David R. Hill
- Abbott Nutrition Research & Development Center, Columbus, OH, United States
| | - Xiaoying Xiong
- Abbott Nutrition Research & Development Center, Shanghai, China
| | - Xiang Li
- Abbott Nutrition Research & Development Center, Shanghai, China
| | - Yanrong Zhao
- Abbott Nutrition Research & Development Center, Shanghai, China
| | - Shuo Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin, China
| |
Collapse
|
43
|
Larke JA, Heiss BE, Ehrlich AM, Taft DH, Raybould HE, Mills DA, Slupsky CM. Milk oligosaccharide-driven persistence of Bifidobacterium pseudocatenulatum modulates local and systemic microbial metabolites upon synbiotic treatment in conventionally colonized mice. MICROBIOME 2023; 11:194. [PMID: 37635250 PMCID: PMC10463478 DOI: 10.1186/s40168-023-01624-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 07/14/2023] [Indexed: 08/29/2023]
Abstract
BACKGROUND Bifidobacteria represent an important gut commensal in humans, particularly during initial microbiome assembly in the first year of life. Enrichment of Bifidobacterium is mediated though the utilization of human milk oligosaccharides (HMOs), as several human-adapted species have dedicated genomic loci for transport and metabolism of these glycans. This results in the release of fermentation products into the gut lumen which may offer physiological benefits to the host. Synbiotic pairing of probiotic species with a cognate prebiotic delivers a competitive advantage, as the prebiotic provides a nutrient niche. METHODS To determine the fitness advantage and metabolic characteristics of an HMO-catabolizing Bifidobacterium strain in the presence or absence of 2'-fucosyllactose (2'-FL), conventionally colonized mice were gavaged with either Bifidobacterium pseudocatenulatum MP80 (B.p. MP80) (as the probiotic) or saline during the first 3 days of the experiment and received water or water containing 2'-FL (as the prebiotic) throughout the study. RESULTS 16S rRNA gene sequencing revealed that mice provided only B.p. MP80 were observed to have a similar microbiota composition as control mice throughout the experiment with a consistently low proportion of Bifidobacteriaceae present. Using 1H NMR spectroscopy, similar metabolic profiles of gut luminal contents and serum were observed between the control and B.p. MP80 group. Conversely, synbiotic supplemented mice exhibited dramatic shifts in their community structure across time with an overall increased, yet variable, proportion of Bifidobacteriaceae following oral inoculation. Parsing the synbiotic group into high and moderate bifidobacterial persistence based on the median proportion of Bifidobacteriaceae, significant differences in gut microbial diversity and metabolite profiles were observed. Notably, metabolites associated with the fermentation of 2'-FL by bifidobacteria were significantly greater in mice with a high proportion of Bifidobacteriaceae in the gut suggesting metabolite production scales with population density. Moreover, 1,2-propanediol, a fucose fermentation product, was only observed in the liver and brain of mice harboring high proportions of Bifidobacteriaceae. CONCLUSIONS This study reinforces that the colonization of the gut with a commensal microorganism does not guarantee a specific functional output. Video Abstract.
Collapse
Affiliation(s)
- Jules A Larke
- Department of Nutrition, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Britta E Heiss
- Department of Food Science and Technology, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Amy M Ehrlich
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California, Davis, , Davis, CA, USA
| | - Diana H Taft
- Department of Food Science and Technology, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Helen E Raybould
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California, Davis, , Davis, CA, USA
| | - David A Mills
- Department of Food Science and Technology, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA.
| | - Carolyn M Slupsky
- Department of Nutrition, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA.
- Department of Food Science and Technology, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA.
| |
Collapse
|
44
|
Pantazi AC, Balasa AL, Mihai CM, Chisnoiu T, Lupu VV, Kassim MAK, Mihai L, Frecus CE, Chirila SI, Lupu A, Andrusca A, Ionescu C, Cuzic V, Cambrea SC. Development of Gut Microbiota in the First 1000 Days after Birth and Potential Interventions. Nutrients 2023; 15:3647. [PMID: 37630837 PMCID: PMC10457741 DOI: 10.3390/nu15163647] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/13/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
The first 1000 days after birth represent a critical window for gut microbiome development, which is essential for immune system maturation and overall health. The gut microbiome undergoes major changes during this period due to shifts in diet and environment. Disruptions to the microbiota early in life can have lasting health effects, including increased risks of inflammatory disorders, autoimmune diseases, neurological disorders, and obesity. Maternal and environmental factors during pregnancy and infancy shape the infant gut microbiota. In this article, we will review how maintaining a healthy gut microbiome in pregnancy and infancy is important for long-term infant health. Furthermore, we briefly include fungal colonization and its effects on the host immune function, which are discussed as part of gut microbiome ecosystem. Additionally, we will describe how potential approaches such as hydrogels enriched with prebiotics and probiotics, gut microbiota transplantation (GMT) during pregnancy, age-specific microbial ecosystem therapeutics, and CRISPR therapies targeting the gut microbiota hold potential for advancing research and development. Nevertheless, thorough evaluation of their safety, effectiveness, and lasting impacts is crucial prior to their application in clinical approach. The article emphasizes the need for continued research to optimize gut microbiota and immune system development through targeted early-life interventions.
Collapse
Affiliation(s)
- Alexandru Cosmin Pantazi
- Pediatrics Department, Faculty of Medicine, “Ovidius” University, 900470 Constanta, Romania (A.L.B.)
- Pediatrics Department, County Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | - Adriana Luminita Balasa
- Pediatrics Department, Faculty of Medicine, “Ovidius” University, 900470 Constanta, Romania (A.L.B.)
- Pediatrics Department, County Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | - Cristina Maria Mihai
- Pediatrics Department, Faculty of Medicine, “Ovidius” University, 900470 Constanta, Romania (A.L.B.)
- Pediatrics Department, County Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | - Tatiana Chisnoiu
- Pediatrics Department, Faculty of Medicine, “Ovidius” University, 900470 Constanta, Romania (A.L.B.)
- Pediatrics Department, County Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | - Vasile Valeriu Lupu
- Pediatrics Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | | | - Larisia Mihai
- Pediatrics Department, Faculty of Medicine, “Ovidius” University, 900470 Constanta, Romania (A.L.B.)
- Pediatrics Department, County Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | - Corina Elena Frecus
- Pediatrics Department, Faculty of Medicine, “Ovidius” University, 900470 Constanta, Romania (A.L.B.)
- Pediatrics Department, County Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | | | - Ancuta Lupu
- Pediatrics Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Antonio Andrusca
- Pediatrics Department, Faculty of Medicine, “Ovidius” University, 900470 Constanta, Romania (A.L.B.)
- Pediatrics Department, County Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | - Constantin Ionescu
- Faculty of Medicine, “Ovidius” University, 900470 Constanta, Romania (S.I.C.)
| | - Viviana Cuzic
- Pediatrics Department, Faculty of Medicine, “Ovidius” University, 900470 Constanta, Romania (A.L.B.)
- Pediatrics Department, County Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | - Simona Claudia Cambrea
- Infectious Diseases Department, Faculty of Medicine, “Ovidius” University, 900470 Constanta, Romania
| |
Collapse
|
45
|
Cho S, Samuel TM, Li T, Howell BR, Baluyot K, Hazlett HC, Elison JT, Zhu H, Hauser J, Sprenger N, Lin W. Interactions between Bifidobacterium and Bacteroides and human milk oligosaccharides and their associations with infant cognition. Front Nutr 2023; 10:1216327. [PMID: 37457984 PMCID: PMC10345227 DOI: 10.3389/fnut.2023.1216327] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023] Open
Abstract
While ample research on independent associations between infant cognition and gut microbiota composition and human milk (HM) oligosaccharides (HMOs) has been reported, studies on how the interactions between gut microbiota and HMOs may yield associations with cognitive development in infancy are lacking. We aimed to determine how HMOs and species of Bacteroides and Bifidobacterium genera interact with each other and their associations with cognitive development in typically developing infants. A total of 105 mother-infant dyads were included in this study. The enrolled infants [2.9-12 months old (8.09 ± 2.48)] were at least predominantly breastfed at 4 months old. A total of 170 HM samples from the mothers and fecal samples of the children were collected longitudinally. Using the Mullen Scales of Early Learning to assess cognition and the scores as the outcomes, linear mixed effects models including both the levels of eight HMOs and relative abundance of Bacteroides and Bifidobacterium species as main associations and their interactions were employed with adjusting covariates; infant sex, delivery mode, maternal education, site, and batch effects of HMOs. Additionally, regression models stratifying infants based on the A-tetrasaccharide (A-tetra) status of the HM they received were also employed to determine if the associations depend on the A-tetra status. With Bacteroides species, we observed significant associations with motor functions, while Bif. catenulatum showed a negative association with visual reception in the detectable A-tetra group both as main effect (value of p = 0.012) and in interaction with LNFP-I (value of p = 0.007). Additionally, 3-FL showed a positive association with gross motor (p = 0.027) and visual reception (p = 0.041). Furthermore, significant associations were observed with the interaction terms mainly in the undetectable A-tetra group. Specifically, we observed negative associations for Bifidobacterium species and LNT [breve (p = 0.011) and longum (p = 0.022)], and positive associations for expressive language with 3'-SL and Bif. bifidum (p = 0.01), 6'-SL and B. fragilis (p = 0.019), and LNFP-I and Bif. kashiwanohense (p = 0.048), respectively. Our findings suggest that gut microbiota and HMOs are both independently and interactively associated with early cognitive development. In particular, the diverse interactions between HMOs and Bacteroides and Bifidobacterium species reveal different candidate pathways through which HMOs, Bifidobacterium and Bacteroides species potentially interact to impact cognitive development in infancy.
Collapse
Affiliation(s)
- Seoyoon Cho
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Tinu M. Samuel
- Nestle Product Technology Center-Nutrition, Société des Produits Nestlé S.A., Vevey, Switzerland
| | - Tengfei Li
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Brittany R. Howell
- Fralin Biomedical Research Institute at VTC, Department of Human Development and Family Science, Virginia Polytechnic Institute and State University, Roanoke, VA, United States
| | - Kristine Baluyot
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Heather C. Hazlett
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Jed T. Elison
- Institute of Child Development, University of Minnesota, Minneapolis, MN, United States
| | - Hongtu Zhu
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Jonas Hauser
- Nestlé Institute of Health Sciences, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Norbert Sprenger
- Nestlé Institute of Health Sciences, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Weili Lin
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
46
|
Fusco W, Lorenzo MB, Cintoni M, Porcari S, Rinninella E, Kaitsas F, Lener E, Mele MC, Gasbarrini A, Collado MC, Cammarota G, Ianiro G. Short-Chain Fatty-Acid-Producing Bacteria: Key Components of the Human Gut Microbiota. Nutrients 2023; 15:2211. [PMID: 37432351 DOI: 10.3390/nu15092211] [Citation(s) in RCA: 281] [Impact Index Per Article: 140.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 04/30/2023] [Accepted: 05/02/2023] [Indexed: 07/12/2023] Open
Abstract
Short-chain fatty acids (SCFAs) play a key role in health and disease, as they regulate gut homeostasis and their deficiency is involved in the pathogenesis of several disorders, including inflammatory bowel diseases, colorectal cancer, and cardiometabolic disorders. SCFAs are metabolites of specific bacterial taxa of the human gut microbiota, and their production is influenced by specific foods or food supplements, mainly prebiotics, by the direct fostering of these taxa. This Review provides an overview of SCFAs' roles and functions, and of SCFA-producing bacteria, from their microbiological characteristics and taxonomy to the biochemical process that lead to the release of SCFAs. Moreover, we will describe the potential therapeutic approaches to boost the levels of SCFAs in the human gut and treat different related diseases.
Collapse
Affiliation(s)
- William Fusco
- Department of Medical and Surgical Sciences, Digestive Disease Center, Universitary Policlinic Agostino Gemelli Foundation IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Manuel Bernabeu Lorenzo
- Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), 46022 Valencia, Spain
| | - Marco Cintoni
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, Universitary Policlinic Agostino Gemelli Foundation IRCCS, 00168 Rome, Italy
| | - Serena Porcari
- Department of Medical and Surgical Sciences, Digestive Disease Center, Universitary Policlinic Agostino Gemelli Foundation IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Emanuele Rinninella
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, Universitary Policlinic Agostino Gemelli Foundation IRCCS, 00168 Rome, Italy
| | - Francesco Kaitsas
- Department of Medical and Surgical Sciences, Digestive Disease Center, Universitary Policlinic Agostino Gemelli Foundation IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Elena Lener
- Department of Medical and Surgical Sciences, Digestive Disease Center, Universitary Policlinic Agostino Gemelli Foundation IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Maria Cristina Mele
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, Universitary Policlinic Agostino Gemelli Foundation IRCCS, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Department of Medical and Surgical Sciences, Digestive Disease Center, Universitary Policlinic Agostino Gemelli Foundation IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), 46022 Valencia, Spain
| | - Giovanni Cammarota
- Department of Medical and Surgical Sciences, Digestive Disease Center, Universitary Policlinic Agostino Gemelli Foundation IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Gianluca Ianiro
- Department of Medical and Surgical Sciences, Digestive Disease Center, Universitary Policlinic Agostino Gemelli Foundation IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| |
Collapse
|
47
|
Abstract
Sialic acids (Sias), a group of over 50 structurally distinct acidic saccharides on the surface of all vertebrate cells, are neuraminic acid derivatives. They serve as glycan chain terminators in extracellular glycolipids and glycoproteins. In particular, Sias have significant implications in cell-to-cell as well as host-to-pathogen interactions and participate in various biological processes, including neurodevelopment, neurodegeneration, fertilization, and tumor migration. However, Sia is also present in some of our daily diets, particularly in conjugated form (sialoglycans), such as those in edible bird's nest, red meats, breast milk, bovine milk, and eggs. Among them, breast milk, especially colostrum, contains a high concentration of sialylated oligosaccharides. Numerous reviews have concentrated on the physiological function of Sia as a cellular component of the body and its relationship with the occurrence of diseases. However, the consumption of Sias through dietary sources exerts significant influence on human health, possibly by modulating the gut microbiota's composition and metabolism. In this review, we summarize the distribution, structure, and biological function of particular Sia-rich diets, including human milk, bovine milk, red meat, and egg.
Collapse
Affiliation(s)
- Tiantian Zhang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Jianrong Wu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Xiaobei Zhan
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| |
Collapse
|
48
|
Willemsen Y, Beijers R, Gu F, Vasquez AA, Schols HA, de Weerth C. Fucosylated Human Milk Oligosaccharides during the First 12 Postnatal Weeks Are Associated with Better Executive Functions in Toddlers. Nutrients 2023; 15:nu15061463. [PMID: 36986193 PMCID: PMC10057664 DOI: 10.3390/nu15061463] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/08/2023] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
Human milk oligosaccharides (HMOs) are one of the most abundant solid components in a mother's milk. Animal studies have confirmed a link between early life exposure to HMOs and better cognitive outcomes in the offspring. Human studies on HMOs and associations with later child cognition are scarce. In this preregistered longitudinal study, we investigated whether human milk 2'-fucosyllactose, 3'-sialyllactose, 6'-sialyllactose, grouped fucosylated HMOs, and grouped sialylated HMOs, assessed during the first twelve postnatal weeks, are associated with better child executive functions at age three years. At infant age two, six, and twelve weeks, a sample of human milk was collected by mothers who were exclusively (n = 45) or partially breastfeeding (n = 18). HMO composition was analysed by use of porous graphitized carbon-ultra high-performance liquid chromatography-mass spectrometry. Executive functions were assessed at age three years with two executive function questionnaires independently filled in by mothers and their partners, and four behavioural tasks. Multiple regression analyses were performed in R. Results indicated that concentrations of 2'-fucosyllactose and grouped fucosylated HMOs were associated with better executive functions, while concentrations of grouped sialylated HMOs were associated with worse executive functions at age three years. Future studies on HMOs that sample frequently during the first months of life and experimental HMO administration studies in exclusively formula-fed infants can further reveal associations with child cognitive development and uncover potential causality and sensitive periods.
Collapse
Affiliation(s)
- Yvonne Willemsen
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 EN Nijmegen, The Netherlands
| | - Roseriet Beijers
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 EN Nijmegen, The Netherlands
- Behavioural Science Institute, Radboud University, 6525 GD Nijmegen, The Netherlands
| | - Fangjie Gu
- Laboratory of Food Chemistry, Wageningen University & Research, 6708 WG Wageningen, The Netherlands
| | - Alejandro Arias Vasquez
- Donders Center for Medical Neuroscience, Department of Psychiatry and Human Genetics, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Henk Arie Schols
- Laboratory of Food Chemistry, Wageningen University & Research, 6708 WG Wageningen, The Netherlands
| | - Carolina de Weerth
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 EN Nijmegen, The Netherlands
| |
Collapse
|
49
|
Hiraku A, Nakata S, Murata M, Xu C, Mutoh N, Arai S, Odamaki T, Iwabuchi N, Tanaka M, Tsuno T, Nakamura M. Early Probiotic Supplementation of Healthy Term Infants with Bifidobacterium longum subsp. infantis M-63 Is Safe and Leads to the Development of Bifidobacterium-Predominant Gut Microbiota: A Double-Blind, Placebo-Controlled Trial. Nutrients 2023; 15:1402. [PMID: 36986131 PMCID: PMC10055625 DOI: 10.3390/nu15061402] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 02/25/2023] [Accepted: 03/02/2023] [Indexed: 03/17/2023] Open
Abstract
Bifidobacteria are important intestinal bacteria that provide a variety of health benefits in infants. We investigated the efficacy and safety of Bifidobacterium longum subsp. infantis (B. infantis) M-63 in healthy infants in a double-blind, randomized, placebo-controlled trial. Healthy term infants were given B. infantis M-63 (n = 56; 1 × 109 CFU/day) or placebo (n = 54) from postnatal age ≤ 7 days to 3 months. Fecal samples were collected, and fecal microbiota, stool pH, short-chain fatty acids, and immune substances were analyzed. Supplementation with B. infantis M-63 significantly increased the relative abundance of Bifidobacterium compared with the placebo group, with a positive correlation with the frequency of breastfeeding. Supplementation with B. infantis M-63 led to decreased stool pH and increased levels of acetic acid and IgA in the stool at 1 month of age compared with the placebo group. There was a decreased frequency of defecation and watery stools in the probiotic group. No adverse events related to test foods were observed. These results indicate that early supplementation with B. infantis M-63 is well tolerated and contributes to the development of Bifidobacterium-predominant gut microbiota during a critical developmental phase in term infants.
Collapse
Affiliation(s)
- Akari Hiraku
- Food Ingredients and Technology Institute, R & D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama 252-8583, Japan
| | - Setsuko Nakata
- Department of Pediatrics, Matsumoto City Hospital, 4417-180, Hata, Matsumoto 390-1401, Japan
| | - Mai Murata
- Food Ingredients and Technology Institute, R & D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama 252-8583, Japan
| | - Chendong Xu
- Food Ingredients and Technology Institute, R & D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama 252-8583, Japan
| | - Natsumi Mutoh
- Food Ingredients and Technology Institute, R & D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama 252-8583, Japan
| | - Satoshi Arai
- Food Ingredients and Technology Institute, R & D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama 252-8583, Japan
| | - Toshitaka Odamaki
- Next Generation Science Institute, R & D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama 252-8583, Japan
| | - Noriyuki Iwabuchi
- Food Ingredients and Technology Institute, R & D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama 252-8583, Japan
| | - Miyuki Tanaka
- Food Ingredients and Technology Institute, R & D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama 252-8583, Japan
| | - Takahisa Tsuno
- Department of Pediatrics, Matsumoto City Hospital, 4417-180, Hata, Matsumoto 390-1401, Japan
| | - Masahiko Nakamura
- Department of neurosurgery, Matsumoto City Hospital, 4417-180, Hata, Matsumoto 390-1401, Japan
| |
Collapse
|
50
|
Infant Fecal Fermentations with Galacto-Oligosaccharides and 2′-Fucosyllactose Show Differential Bifidobacterium longum Stimulation at Subspecies Level. CHILDREN 2023; 10:children10030430. [PMID: 36979988 PMCID: PMC10047592 DOI: 10.3390/children10030430] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/26/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023]
Abstract
The objective of the current study was to evaluate the potential of 2′-FL and GOS, individually and combined, in beneficially modulating the microbial composition of infant and toddler (12–18 months) feces using the micro-Matrix bioreactor. In addition, the impacts of GOS and 2′-FL, individually and combined, on the outgrowth of fecal bifidobacteria at (sub)species level was investigated using the baby M-SHIME® model. For young toddlers, significant increases in the genera Bifidobacterium, Veillonella, and Streptococcus, and decreases in Enterobacteriaceae, Clostridium XIVa, and Roseburia were observed in all supplemented fermentations. In addition, GOS, and combinations of GOS and 2′-FL, increased Collinsella and decreased Salmonella, whereas 2′-FL, and combined GOS and 2′-FL, decreased Dorea. Alpha diversity increased significantly in infants with GOS and/or 2′-FL, as well as the relative abundances of the genera Veillonella and Akkermansia with 2′-FL, and Lactobacillus with GOS. Combinations of GOS and 2′-FL significantly stimulated Veillonella, Lactobacillus, Bifidobacterium, and Streptococcus. In all supplemented fermentations, Proteobacteria decreased, with the most profound decreases accomplished by the combination of GOS and 2′-FL. When zooming in on the different (sub)species of Bifidobacterium, GOS and 2’-FL were shown to be complementary in stimulating breast-fed infant-associated subspecies of Bifidobacterium longum in a dose-dependent manner: GOS stimulated Bifidobacterium longum subsp. longum, whereas 2′-FL supported outgrowth of Bifidobacterium longum subsp. infantis.
Collapse
|