1
|
Nakajima A, Arzamasov AA, Sakanaka M, Murakami R, Kozakai T, Yoshida K, Katoh T, Ojima MN, Hirose J, Nagao S, Xiao JZ, Odamaki T, Rodionov DA, Katayama T. In vitro competition with Bifidobacterium strains impairs potentially pathogenic growth of Clostridium perfringens on 2'-fucosyllactose. Gut Microbes 2025; 17:2478306. [PMID: 40102238 PMCID: PMC11956901 DOI: 10.1080/19490976.2025.2478306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 02/23/2025] [Accepted: 03/04/2025] [Indexed: 03/20/2025] Open
Abstract
Fortifying infant formula with human milk oligosaccharides, such as 2'-fucosyllactose (2'-FL), is a global trend. Previous studies have shown the inability of pathogenic gut microbes to utilize 2'-FL. However, the present study demonstrates that the type strain (JCM 1290T) of Clostridium perfringens, a pathobiont species often more prevalent and abundant in the feces of C-section-delivered infants, exhibits potentially pathogenic growth on 2'-FL. The expression of genes for α-toxin, an activator of NLRP3 inflammasome, and ethanolamine ammonia-lyase, a factor responsible for the progression of gas gangrene, was significantly upregulated during 2'-FL assimilation compared to growth on lactose. However, colony-forming unit of C. perfringens JCM 1290T markedly decreased when co-cultivated with selected strains of Bifidobacterium, a taxon frequently detected in the breastfed infant gut. Moreover, during co-cultivation, the expression of virulence-related genes, including the gene for perfringolysin O - another activator of NLRP3 inflammasome - were significantly downregulated, while the lactate oxidation genes were upregulated. This can occur through two different mechanisms: direct competition for 2'-FL between the two organisms, or cross-feeding of lactose, released from 2'-FL by C. perfringens JCM 1290T, to Bifidobacterium. Attenuation of α-toxin production by the selected Bifidobacterium strains was observed to varying extents in 2'-FL-utilizing C. perfringens strains clinically isolated from healthy infants. Our results warrant detailed in vivo studies using animal models with dysbiotic microbiota dominated by various types of C. perfringens strains to further validate the safety of 2'-FL for clinical interventions, particularly on vulnerable preterm infants.
Collapse
Affiliation(s)
- Aruto Nakajima
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Aleksandr A. Arzamasov
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | | | - Ryuta Murakami
- Innovative Research Institute, Morinaga Milk Industry Co, Ltd, Zama, Kanagawa, Japan
| | - Tomoya Kozakai
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Keisuke Yoshida
- Innovative Research Institute, Morinaga Milk Industry Co, Ltd, Zama, Kanagawa, Japan
| | - Toshihiko Katoh
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Miriam N. Ojima
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Junko Hirose
- Department of Food and Nutrition, Kyoto Women’s University, Kyoto, Japan
| | | | - Jin-Zhong Xiao
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Innovative Research Institute, Morinaga Milk Industry Co, Ltd, Zama, Kanagawa, Japan
| | - Toshitaka Odamaki
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Innovative Research Institute, Morinaga Milk Industry Co, Ltd, Zama, Kanagawa, Japan
| | - Dmitry A. Rodionov
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Takane Katayama
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| |
Collapse
|
2
|
Huang R, Zhou G, Cai J, Cao C, Zhu Z, Wu Q, Zhang F, Ding Y. Maternal consumption of urbanized diet compromises early-life health in association with gut microbiota. Gut Microbes 2025; 17:2483783. [PMID: 40176259 PMCID: PMC11988223 DOI: 10.1080/19490976.2025.2483783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/09/2025] [Accepted: 03/18/2025] [Indexed: 04/04/2025] Open
Abstract
Urbanization has significantly transformed dietary habits worldwide, contributing to a globally increased burden of non-communicable diseases and altered gut microbiota landscape. However, it is often overlooked that the adverse effects of these dietary changes can be transmitted from the mother to offspring during early developmental stages, subsequently influencing the predisposition to various diseases later in life. This review aims to delineate the detrimental effects of maternal urban-lifestyle diet (urbanized diet) on early-life health and gut microbiota assembly, provide mechanistic insights on how urbanized diet mediates mother-to-offspring transfer of bioactive substances in both intrauterine and extrauterine and thus affects fetal and neonatal development. Moreover, we also further propose a framework for developing microbiome-targeted precision nutrition and diet strategies specifically for pregnant and lactating women. The establishment of such knowledge can help develop proactive preventive measures from the beginning of life, ultimately reducing the long-term risk of disease and improving public health outcomes.
Collapse
Affiliation(s)
- Rong Huang
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Guicheng Zhou
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Jie Cai
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Cha Cao
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Zhenjun Zhu
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Qingping Wu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Fen Zhang
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Yu Ding
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| |
Collapse
|
3
|
Christensen KR, Rasmussen TS, Mentzel CMJ, Lanng SK, Meloni ETG, Bertram HC, Hansen CHF, Hansen AK. The Impact of Human Milk Oligosaccharides on Antibiotic-Induced Microbial Dysbiosis and Gut Inflammation in Mice. Antibiotics (Basel) 2025; 14:488. [PMID: 40426555 PMCID: PMC12108310 DOI: 10.3390/antibiotics14050488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 05/05/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: Antibiotics have a significant impact on the gut microbiota, and we hypothesized that human milk oligosaccharides may alleviate antibiotic-induced gut microbiota dysbiosis. Methods: Six groups of eight mice were administered drinking water with or without ampicillin for one week. We then introduced the human milk oligosaccharide 2'-fucosyllactose (2'FL), either alone or in combination with difucosyl-lactose (DFL), for two weeks after the termination of ampicillin treatment. Results: Ampicillin reduced microbiota diversity and the abundance of specific bacteria. One week after the termination of ampicillin treatment, the 2'FL + DFL mixture counteracted the ampicillin-induced reduction in diversity, although this effect was not sustained. Over the subsequent two weeks, the 2'FL + DFL mixture had a significant impact on the relative abundances of Lactobacillus spp. and Bacteroides spp. Ampicillin also reduced caecal propionate levels, downregulated the gene Gzmb for Granzyme B, and upregulated the gene Reg3a for Regenerating islet-derived protein 3 alpha, all of which were counteracted by the 2'FL + DFL mixture. Ampicillin had a minor impact on ileal cytokine levels. The 2'FL + DFL mixture showed a cytokine effect indicating reduced adaptive and innate inflammation. Ampicillin reduced water intake and growth in the mice. The oligosaccharides did not affect water intake, but the 2'FL + DFL mixture slightly reduced body weight. Conclusions: The 2'FL + DFL mixture appears to hold potential for counteracting some of the side effects of ampicillin treatment.
Collapse
Affiliation(s)
- Kristine Rothaus Christensen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Ridebanevej 9, DK-1870 Frederiksberg C, Denmark; (K.R.C.); (C.M.J.M.); (C.H.F.H.)
- Dsm-Firmenich, Kogle Allé 4, DK-2970 Hørsholm, Denmark
| | - Torben Sølbeck Rasmussen
- Department of Food Science, University of Copenhagen, Rolighedsvej 26, DK-1958 Frederiksberg C, Denmark; (T.S.R.); (E.T.G.M.)
| | - Caroline M. Junker Mentzel
- Department of Veterinary and Animal Sciences, University of Copenhagen, Ridebanevej 9, DK-1870 Frederiksberg C, Denmark; (K.R.C.); (C.M.J.M.); (C.H.F.H.)
| | - Sofie Kaas Lanng
- Department of Food Science, Aarhus University, Agro Food Park 48, DK-8200 Aarhus N, Denmark; (S.K.L.); (H.C.B.)
| | - Elena Tina Gabriella Meloni
- Department of Food Science, University of Copenhagen, Rolighedsvej 26, DK-1958 Frederiksberg C, Denmark; (T.S.R.); (E.T.G.M.)
- Department of Biotechnology and Biosciences BtBs, Piazza dell’Ateneo Nuovo, I-1-20126 Milan, Italy
| | - Hanne Christine Bertram
- Department of Food Science, Aarhus University, Agro Food Park 48, DK-8200 Aarhus N, Denmark; (S.K.L.); (H.C.B.)
| | - Camilla Hartmann Friis Hansen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Ridebanevej 9, DK-1870 Frederiksberg C, Denmark; (K.R.C.); (C.M.J.M.); (C.H.F.H.)
| | - Axel Kornerup Hansen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Ridebanevej 9, DK-1870 Frederiksberg C, Denmark; (K.R.C.); (C.M.J.M.); (C.H.F.H.)
| |
Collapse
|
4
|
Häsler R, Mikš MH, Bajic D, Soyyilmaz B, Bendik I, van Buul VJ, Steinert RE, Rehman A. Human Milk Oligosaccharides Modulating Inflammation in Infants, Adults, and Older Individuals-From Concepts to Applications. Adv Nutr 2025; 16:100433. [PMID: 40287068 DOI: 10.1016/j.advnut.2025.100433] [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: 09/16/2024] [Revised: 04/17/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025] Open
Abstract
The increasing global prevalence of inflammatory diseases, such as ulcerative colitis and irritable bowel syndrome, represents a challenging task for healthcare systems. Several approaches to disease management target the intestinal microbiome, which plays a key role in health and disease. One promising approach is modulating the microbiome using human milk oligosaccharides (HMOs). Originating from human milk, HMOs are indigestible carbohydrates that act in a host-optimized prebiotic fashion by providing an energy source for health-promoting intestinal bacteria and exhibiting systemic effects. Commercial products supporting infant health and development have been the primary fields of HMO application. Advancements in the large-scale production of HMOs through bioengineering and precision fermentation have led to evaluation of their potential for managing inflammatory diseases. Several in vitro studies and observations on model systems have been clinically validated in infants, resulting in a large body of evidence supporting the safety and efficacy of HMOs in inflammatory disorders. Although novel approaches seek to explore interventions in adults, the primary goal for the future is to provide cost-efficient, safe, and reliable healthcare compounds across all age groups.
Collapse
Affiliation(s)
- Robert Häsler
- Department of Dermatology and Allergology, University Kiel, Kiel, Germany.
| | - Marta Hanna Mikš
- Faculty of Food Science, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland; dsm-firmenich, Glycom A/S, Hørsholm, Denmark
| | - Danica Bajic
- dsm-firmenich, Health, Nutrition & Care, Kaiseraugst, Switzerland
| | | | - Igor Bendik
- dsm-firmenich, Health, Nutrition & Care, Kaiseraugst, Switzerland
| | | | | | - Ateequr Rehman
- dsm-firmenich, Health, Nutrition & Care, Kaiseraugst, Switzerland
| |
Collapse
|
5
|
Liu S, Zeng X, Li J, Li W, Gu Y, Li B, Wang J. Goat milk oligosaccharides: regulating infant immunity by intervention in the gut microbiota. Food Funct 2025; 16:2213-2229. [PMID: 40035489 DOI: 10.1039/d5fo00162e] [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: 03/05/2025]
Abstract
The health status of the growing infant is closely related to the development of the gut microbiota during infancy, which is also a major stimulator of the immune system. Goat milk oligosaccharides (gMOs) are a class of bioactive compounds in goat milk, which have attracted extensive research interest in recent years. Recent studies have highlighted that gMOs as prebiotics can regulate the gut microbiota, exhibit multiple health effects, and act as immunomodulators. This article outlines the structure, classification, and functions of gMOs. In addition, we also deeply explored the mechanism of gMO interaction with infant gut microbiota and regulation of infant immunity. Finally, the possibility of gMOs as an effective substitute for natural prebiotics in breast milk is revisited. We concluded that gMOs improve infant immune function by regulating intestinal beneficial bacteria (Bifidobacteria, Lactobacilli, etc.) and their metabolism. Therefore, gMOs are significant to infant immune health and are expected to become a substitute for human milk oligosaccharides (HMOs).
Collapse
Affiliation(s)
- Sibo Liu
- Food College, Northeast Agricultural University, Harbin 150030, China.
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China
| | - Xiaoling Zeng
- Ausnutria Dairy (China) Co., Ltd, Changsha 410000, China.
| | - Jing Li
- Ausnutria Dairy (China) Co., Ltd, Changsha 410000, China.
| | - Wei Li
- Ausnutria Dairy (China) Co., Ltd, Changsha 410000, China.
| | - Yue Gu
- Food College, Northeast Agricultural University, Harbin 150030, China.
| | - Bailiang Li
- Food College, Northeast Agricultural University, Harbin 150030, China.
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China
| | - Jiaqi Wang
- Ausnutria Dairy (China) Co., Ltd, Changsha 410000, China.
| |
Collapse
|
6
|
Lazarini T, Tonon KM, de Araujo Filho HB, de Morais MB. Bifidogenic Effect of 2'-Fucosyllactose (2'-FL) on the Gut Microbiome of Healthy Formula-Fed Infants: A Randomized Clinical Trial. Nutrients 2025; 17:973. [PMID: 40290019 PMCID: PMC11944528 DOI: 10.3390/nu17060973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 02/28/2025] [Accepted: 03/05/2025] [Indexed: 04/30/2025] Open
Abstract
Breast milk is rich in bioactive components, especially human milk oligosaccharides (HMOs), which are crucial for establishing gut microbiota. The 2'-FL (2-Fucosyllactose), one of the most abundant oligosaccharides in breast milk, functions as a selective prebiotic. Objective: To examine the effect of adding 2'-FL (2-Fucosyllactose) to an infant formula containing prebiotic galacto-oligosaccharides (GOSs) and fructo-oligosaccharides (FOSs) on the gut microbiome of healthy formula-fed infants. Methods: This study enrolled infants from three groups: an HMO experimental group (n = 29), a GOS/FOS control group (n = 30), and an exclusively breastfed (breast milk [BM]) reference group (n = 28). Fecal samples from the three groups in the first and fourth months of life were analyzed. The V3 and V4 regions of the 16S rRNA gene were amplified and sequenced on the Illumina MiSeq. ANOVA, Kruskal-Wallis, richness indices (Chao1, Shannon), UniFrac distances, and the Adonis tests were used to perform statistical analyses on the relative abundance of phyla and genera, as well as the alpha and beta-diversity of the gut microbiota. Results: After intervention, Actinobacteriota emerged as the predominant phylum in both the HMO (60.4%) and BM (46.6%) groups. Bifidobacterium and Escherichia-Shigella were identified as the two most abundant bacterial genera in both groups. Nevertheless, the statistical analysis showed that the relative abundance of Bifidobacterium in the HMO formula-fed group after intervention was similar to that in the BM group (p > 0.05). Infants in the HMO and GOS/FOS groups showed higher relative abundance of [Ruminococcus]_gnavus_group bacteria compared to those in the BM group. Groups fed with infant formula demonstrated higher alpha-diversity of gut microbiota compared to breastfed infants (p < 0.05), at the time of admission as well as after the intervention. Beta-diversity was significantly different among the three groups, according to type of feeding. Infants fed a 2'-FL-supplemented infant formula exhibited growth comparable to that of breastfed infants throughout the intervention period, demonstrating that the formula was both safe and well tolerated. Conclusions: Adding 2'-FL to an infant formula containing 4 g/L of GOS + FOS resulted in a stronger bifidogenic effect compared to the formula without 2'-FL.
Collapse
Affiliation(s)
- Tamara Lazarini
- Nutrition Postgraduate Program, Universidade Federal de São Paulo, São Paulo 04023-062, Brazil;
| | - Karina Merini Tonon
- Department of Environmental & Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA;
| | | | - Mauro Batista de Morais
- Nutrition Postgraduate Program, Universidade Federal de São Paulo, São Paulo 04023-062, Brazil;
- Division of Pediatric Gastroenterology, Universidade Federal de São Paulo, São Paulo 04023-062, Brazil;
| |
Collapse
|
7
|
Du J, Yang H. 2'-Fucosyllactose as a prebiotic modulates the probiotic responses of Bifidobacterium bifidum. Curr Res Food Sci 2025; 10:100975. [PMID: 39906504 PMCID: PMC11791163 DOI: 10.1016/j.crfs.2025.100975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/04/2025] [Accepted: 01/14/2025] [Indexed: 02/06/2025] Open
Abstract
2'-Fucosyllactose (2'-FL), one of the most representative oligosaccharides in human milk, is intimately linked to the enrichment of specific Bifidobacterium species. However, the efficacy of 2'-FL in modulating the probiotic responses of bifidobacterium has been rarely researched. Thereinto, three key issues have yet to be reported: the effects of 2'-FL hydrolysis on bifidobacterial growth, the protective effects of 2'-FL on bifidobacterium under gastrointestinal stress and the inhibitory activity of 2'-FL metabolites against Cronobacter spp. This work intended to address these concerns. 2'-FL dramatically accelerated the growth and proliferation of Bifidobacterium bifidum YH17 and Bifidobacterium bifidum BBI01. The glucose in lactose core on 2'-FL was preferable for B. bifidum to achieve substantial increases in biomass while the galactose was not readily available. Additionally, 2'-FL showed unique advantages in ameliorating the resistance of B. bifidum to gastrointestinal challenges. 2'-FL considerably improved the adhesive property of B. bifidum, thus facilitating the competitive elimination of Cronobacter sakazakii ATCC 29544 and Cronobacter muytjensii ATCC 51329 by B. bifidum. The growth inhibition of 2'-FL on the Cronobacter strains was mediated by promoting the secretion of antibacterial substances from B. bifidum. The inhibitory activity hinged on the B. bifidum strains. 2'-FL specifically induced B. bifidum BBI01 to produce some antibacterial substances that were proteinaceous, thermostable and relatively stable even at pH 8.0. These antibacterial substances played a key role in the inhibitory activity and had a synergistic effect with acidification. These observations provide a useful guideline for developing synbiotic supplements to intervene the infant gut microbiota.
Collapse
Affiliation(s)
- Jingfang Du
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Hong Yang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
8
|
Chen G, Chen L, Wang H, Zhang J, Sun X, Chen X, Fan J, Jia Z, Huang Y. 1H Nuclear Magnetic Resonance-Based Metabolomic Profiling and Comparison of Human Milk across Different Lactation Stages in Secretors and Nonsecretors: A Study of Chinese Lactating Women. J Nutr 2025; 155:78-86. [PMID: 39491676 DOI: 10.1016/j.tjnut.2024.10.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/22/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024] Open
Abstract
BACKGROUND Human milk oligosaccharides (HMOs) and other milk-derived metabolites are crucial for infant health, influencing gut microbiota and overall development. OBJECTIVE This study aimed to uncover insights into the variations of HMOs and non-HMO metabolites based on secretor (Se) status, lactation time, mode of delivery, and infant sex. METHODS An exploratory cross-sectional study was designed to compare the concentrations of HMOs and non-HMOs metabolites in milk samples from 129 lactating Chinese women within 1 y postpartum. Nuclear magnetic resonance analysis was employed for the identification and quantification of the metabolites. The metabolites measured were grouped into sugars, free amino acids, fatty acids, and metabolites related to energy metabolism. The influences of delivery mode and infant sex on milk metabolite composition were explored. RESULTS Uniform Manifold Approximation and Projection analysis of HMOs profiles revealed distinct clustering based on Se status, with significant differences in 2'-fucosyllactose (2'-FL) and 3-fucosyllactose (3-FL) concentrations observed between Se+ and Se- groups. A decreasing trend for 2'-FL and 6'-sialyllactose concentrations, along with an increase in 3-FL concentrations, was observed with increasing lactating period within 12 mo postpartum. Non-HMOs metabolite analysis indicated that Se status only affected glutamate concentrations. An increase in glutamine concentrations was observed 3-9 mo postpartum. A continuous increase in o-phosphocholine concentrations was noted in 12 mo postpartum, along with reductions in citrate and sn-glycero-phosphocholine concentrations. Delivery mode and infant sex did not affect both HMOs and non-HMOs concentrations. CONCLUSIONS Metabolomic analysis of human milk reveals significant variation of HMOs, but not in non-HMOs, based on Se status. Changes in certain HMOs and non-HMOs concentrations were also observed over the 1 y of lactation. Understanding how these metabolites change over time may influence recommendations for maternal diet, supplementation, and the timing of breastfeeding to ensure optimal nutrient delivery to the infant.
Collapse
Affiliation(s)
- Guixia Chen
- Department of Child Health, Department of Pediatrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China.
| | - Lifeng Chen
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, United States
| | - Huiya Wang
- Department of Clinic, Department of Pediatrics, Women and Children's Hospital, School of Mecicine, Xiamen University, Xiamen, China
| | - Jiyong Zhang
- Department of Child Health, Department of Pediatrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Xiaoling Sun
- Department of Child Health, Department of Pediatrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Xiaoxin Chen
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States
| | - Jianxia Fan
- Department of Child Health, Department of Pediatrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Zhiwei Jia
- Department of Child Health, Department of Pediatrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Yinying Huang
- Department of Nursing, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
9
|
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
|
10
|
Sato K, Nakamura Y, Fujiyama K, Ohneda K, Nobukuni T, Ogishima S, Mizuno S, Koshiba S, Kuriyama S, Jinno S. Absolute quantification of eight human milk oligosaccharides in breast milk to evaluate their concentration profiles and associations with infants' neurodevelopmental outcomes. J Food Sci 2024; 89:10152-10170. [PMID: 39656795 DOI: 10.1111/1750-3841.17597] [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: 06/21/2024] [Revised: 11/03/2024] [Accepted: 11/21/2024] [Indexed: 12/17/2024]
Abstract
Human milk oligosaccharides (HMOs) have been positively associated with child neurodevelopment in some cohort studies. However, there is a lack of consistency in the association between HMOs and benefits to infants' brains. Moreover, the quantification methods for HMOs have not yet been standardized. In this study, we developed a quantification method for evaluating eight HMOs (2'-fucosyllactose [2'-FL], 3'-fucosyllactose [3'-FL], 3'-sialyllactose [3'-SL], 6'-sialyllactose [6'-SL], lactosialyltetrasaccharide a [LSTa], lactosialyltetrasaccharide b [LSTb], lactosialyltetrasaccharide c [LSTc], and disialyllacto-N-tetraose [DSLNT]) in breast milk. After validating the method, we applied it to 1-month breast milk samples (n = 150) from the Tohoku Medical Megabank Project Birth and Three-Generation Cohort Study to assess HMO profiles in breast milk and their possible association with changes in head circumference z-score (ΔHCZ) and neurodevelopmental scores of children (as measured by the Ages and Stages Questionnaire, Third Edition). The validation demonstrated that the method had relative standard deviation ≤ 12.7% of precision and 79.5-110.9% of accuracy. Using this method, eight HMO levels (2'-FL, 0-4.74 mg/mL; 3'-FL, 0.02-1.52 mg/mL; 3'-SL, 0.07-0.32 mg/mL; 6'-SL, 0.01-0.70 mg/mL; LSTa, 0.002-0.043 mg/mL; LSTb, 0.02-0.31 mg/mL; LSTc, 0.001-0.47 mg/mL; and DSLNT, 0.09-0.71 mg/mL [min-max, all participants]) and the ratio of low secretors (16.0%) in the Japanese cohort were obtained. The obtained HMO levels in breast milk were subjected to multivariate analysis to screen for HMOs showing a positive association with ΔHCZ and neurodevelopmental scores. The results proposed that ΔHCZ was positively associated with LSTb and 2'-FL levels, whereas neurodevelopmental scores were positively associated with 2'-FL levels (among all participants) and 3'-SL and DSLNT levels (among secretor participants). This study showed that the developed method provides HMO profiles in Japanese breast milk, as well as additional information on the associations between specific HMOs and neurodevelopment, reinforcing the sum of evidence for the role of HMOs in the brain.
Collapse
Affiliation(s)
- Keigo Sato
- Food Microbiology and Function Research Laboratory, Meiji Co., Ltd., Hachioji, Japan
- Wellness Science Labs, Meiji Holdings Co., Ltd., Hachioji, Japan
- International Centre for Biotechnology, Osaka University, Suita, Japan
| | - Yoshitaka Nakamura
- Food Microbiology and Function Research Laboratory, Meiji Co., Ltd., Hachioji, Japan
| | - Kazuhito Fujiyama
- International Centre for Biotechnology, Osaka University, Suita, Japan
| | - Kinuko Ohneda
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Takahiro Nobukuni
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Soichi Ogishima
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Japan
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Sendai, Japan
| | - Satoshi Mizuno
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Seizo Koshiba
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Japan
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Sendai, Japan
| | - Shinichi Kuriyama
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Japan
- International Research Institute of Disaster Science, Tohoku University, Sendai, Japan
| | - Shinji Jinno
- Food Microbiology and Function Research Laboratory, Meiji Co., Ltd., Hachioji, Japan
- Wellness Science Labs, Meiji Holdings Co., Ltd., Hachioji, Japan
| |
Collapse
|
11
|
Zhu L, Wang M, Li H, Luo T, Deng Z, Li J, Zheng L, Zhang B. Supplementation of 3'-Sialyllactose During the Growth Period Improves Learning and Memory Development in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:24518-24529. [PMID: 39454104 DOI: 10.1021/acs.jafc.4c06106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2024]
Abstract
3'-Sialyllactose (3'-SL), a major acidic oligosaccharide found in human milk, has been investigated to improve cognitive-enhancing effects with 3 weeks old C57BL/6 mice by administering 3'-SL orally at a dose of 350 mg/kg/day for 6 weeks. Behavioral tests indicated that supplementation with 3'-SL promoted cognitive and memory development in young mice. Through interaction network and coenrichment analysis, nine differentially expressed genes (DEGs) related to memory and cognition were identified and localized in the hippocampal tissue of mice. The intervention of 3'-SL significantly increased the metabolism of sialic acid in mouse hippocampal tissue and promoted the expression of learning-related genes (p < 0.05). Notably, it increased the expression of genes associated with neural cell adhesion molecule (NCAM, p < 0.05), glutamate receptors, and fibroblast growth factor receptor (FGFR, p < 0.05). This suggests that 3'-SL may elevate polysialylated NCAM (PSA-NCAM) levels, which could subsequently interact with FGFR and glutamate receptors, thereby enhancing synaptic growth and plasticity. Additionally, 3'-SL altered the composition of the mouse intestinal microbiota. The synergistic action of gut microbiota and intestinal sialidase promoted the production of free sialic acid, providing essential nutritional elements for the development of the brain's nervous system. In conclusion, our findings provide new insights into the promoting effect of 3'-SL on cognitive development in growing mice and elucidate its molecular mechanisms.
Collapse
Affiliation(s)
- Liuying Zhu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Minghui Wang
- 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
- International Institute of Food Innovation, Nanchang University, Nanchang 330051, Jiangxi, China
| | - Ting Luo
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
- International Institute of Food Innovation, Nanchang University, Nanchang 330051, Jiangxi, China
| | - Zeyuan Deng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
- International Institute of Food Innovation, Nanchang University, Nanchang 330051, Jiangxi, China
| | - Jing Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
- International Institute of Food Innovation, Nanchang University, Nanchang 330051, Jiangxi, China
| | - Liufeng Zheng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
- International Institute of Food Innovation, Nanchang University, Nanchang 330051, Jiangxi, China
| | - Bing Zhang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
- International Institute of Food Innovation, Nanchang University, Nanchang 330051, Jiangxi, China
| |
Collapse
|
12
|
Wichmann A. Biological effects of combinations of structurally diverse human milk oligosaccharides. Front Pediatr 2024; 12:1439612. [PMID: 39564380 PMCID: PMC11573541 DOI: 10.3389/fped.2024.1439612] [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: 05/28/2024] [Accepted: 10/14/2024] [Indexed: 11/21/2024] Open
Abstract
Human milk oligosaccharides (HMOs) are a diverse group of structures and an abundant bioactive component of breastmilk that contribute to infant health and development. Preclinical studies indicate roles for HMOs in shaping the infant gut microbiota, inhibiting pathogens, modulating the immune system, and influencing cognitive development. In the past decade, several industrially produced HMOs have become available to fortify infant formula. Clinical intervention trials with manufactured HMOs have begun to corroborate some of the physiological effects reported in preclinical studies, especially modulation of the gut microbiota in the direction of breastfed infants. As more HMOs become commercially available and as HMOs have some shared mechanisms of action, there is a need to better understand the unique and differential effects of individual HMOs and the benefits of combining multiple HMOs. This review focuses on the differential effects of different HMO structural classes and individual structures and presents a scientific rationale for why combining multiple structurally diverse HMOs is expected to exert greater biological effects.
Collapse
Affiliation(s)
- Anita Wichmann
- Global Regulatory Affairs HMOs, Early Life & Medical Nutrition, DSM-Firmenich, Hørsholm, Denmark
| |
Collapse
|
13
|
Ma X, Li M, Wang X, Qi G, Wei L, Zhang D. Sialylation in the gut: From mucosal protection to disease pathogenesis. Carbohydr Polym 2024; 343:122471. [PMID: 39174097 DOI: 10.1016/j.carbpol.2024.122471] [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: 05/07/2024] [Revised: 06/19/2024] [Accepted: 07/07/2024] [Indexed: 08/24/2024]
Abstract
Sialylation, a crucial post-translational modification of glycoconjugates, entails the attachment of sialic acid (SA) to the terminal glycans of glycoproteins and glycolipids through a tightly regulated enzymatic process involving various enzymes. This review offers a comprehensive exploration of sialylation within the gut, encompassing its involvement in mucosal protection and its impact on disease progression. The sialylation of mucins and epithelial glycoproteins contributes to the integrity of the intestinal mucosal barrier. Furthermore, sialylation regulates immune responses in the gut, shaping interactions among immune cells, as well as their activation and tolerance. Additionally, the gut microbiota and gut-brain axis communication are involved in the role of sialylation in intestinal health. Altered sialylation patterns have been implicated in various intestinal diseases, including inflammatory bowel disease (IBD), colorectal cancer (CRC), and other intestinal disorders. Emerging research underscores sialylation as a promising avenue for diagnostic, prognostic, and therapeutic interventions in intestinal diseases. Potential strategies such as sialic acid supplementation, inhibition of sialidases, immunotherapy targeting sialylated antigens, and modulation of sialyltransferases have been utilized in the treatment of intestinal diseases. Future research directions will focus on elucidating the molecular mechanisms underlying sialylation alterations, identifying sialylation-based biomarkers, and developing targeted interventions for precision medicine approaches.
Collapse
Affiliation(s)
- Xueni Ma
- Key Laboratory of Digestive Diseases, Lanzhou University Second Hospital, Lanzhou, China; The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Muyang Li
- Key Laboratory of Digestive Diseases, Lanzhou University Second Hospital, Lanzhou, China; The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Xiaochun Wang
- Department of Gastroenterology, Gansu Provincial Hospital, Lanzhou, China
| | - Guoqing Qi
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, China
| | - Lina Wei
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, China
| | - Dekui Zhang
- Key Laboratory of Digestive Diseases, Lanzhou University Second Hospital, Lanzhou, China; Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, China.
| |
Collapse
|
14
|
Shi Y, Yin R, Pang J, Chen Y, Li Z, Su S, Wen Y. Impact of complementary feeding on infant gut microbiome, metabolites and early development. Food Funct 2024; 15:10663-10678. [PMID: 39354871 DOI: 10.1039/d4fo03948c] [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: 10/03/2024]
Abstract
Introducing complementary foods is critical for promoting infant health and development. During the weaning period, the dietary patterns provide essential nutrients and facilitate the development of a diverse gut microbiome, which plays significant roles in the regulation of immune, metabolic, and neurological functions. This study enrolled 200 families to assess the impact of complementary feeding on infant growth and health outcomes. Data included detailed records of feeding practices, infant growth measurements, health assessments, and fecal samples and breast milk collected between weeks 12 and 32 postpartum. The gut microbiome was analyzed using 16S rRNA sequencing, while metabolites such as human milk oligosaccharides (HMOs), monosaccharides, and short-chain fatty acids (SCFAs) were measured using chromatography-mass spectrometry. Results revealed a high prevalence of breastfeeding, with complementary food introduced at around 16 weeks. Significant alterations in the infant gut microbiome were observed, particularly in the genera Lactobacillus, Akkermansia, and Staphylococcus. Additionally, the levels of HMOs, monosaccharides, and SCFAs were found to be influenced by the introduction of complementary foods. Significant correlations emerged between complementary feeding practices, gut microbiome diversity, specific bacterial genera (e.g., Streptococcus, Lactobacillus, Bifidobacterium, and Clostridioides), and key metabolites (such as lacto-N-tetraose, lacto-N-neotetraose, mannose, and butyric acid). This study offers valuable insights into the complex interactions between complementary feeding, gut microbiome development, and metabolite profiles during early infant growth. Future research with larger cohorts and targeted dietary interventions is recommended to further elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Yudong Shi
- Global R&D Innovation Center, Inner Mongolia Mengniu Dairy Group Co Ltd, China.
| | - Ran Yin
- Global R&D Innovation Center, Inner Mongolia Mengniu Dairy Group Co Ltd, China.
| | - Jinzhu Pang
- Global R&D Innovation Center, Inner Mongolia Mengniu Dairy Group Co Ltd, China.
| | - Yun Chen
- Global R&D Innovation Center, Inner Mongolia Mengniu Dairy Group Co Ltd, China.
| | - Zhouyong Li
- Global R&D Innovation Center, Inner Mongolia Mengniu Dairy Group Co Ltd, China.
| | - Shengpeng Su
- Global R&D Innovation Center, Inner Mongolia Mengniu Dairy Group Co Ltd, China.
| | - Yongping Wen
- Global R&D Innovation Center, Inner Mongolia Mengniu Dairy Group Co Ltd, China.
| |
Collapse
|
15
|
Du Z, Li Z, Guang C, Zhu Y, Mu W. Recent advances of 3-fucosyllactose in health effects and production. Arch Microbiol 2024; 206:378. [PMID: 39143417 DOI: 10.1007/s00203-024-04104-2] [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: 06/28/2024] [Revised: 07/29/2024] [Accepted: 08/05/2024] [Indexed: 08/16/2024]
Abstract
Human milk oligosaccharides (HMOs) have been recognized as gold standard for infant development. 3-Fucosyllactose (3-FL), being one of the Generally Recognized as Safe HMOs, represents a core trisaccharide within the realm of HMOs; however, it has received comparatively less attention in contrast to extensively studied 2'-fucosyllactose. The objective of this review is to comprehensively summarize the health effects of 3-FL, including its impact on gut microbiota proliferation, antimicrobial effects, immune regulation, antiviral protection, and brain maturation. Additionally, the discussion also covers the commercial application and regulatory approval status of 3-FL. Lastly, an organized presentation of large-scale production methods for 3-FL aims to provide a comprehensive guide that highlights current strategies and challenges in optimization.
Collapse
Affiliation(s)
- Zhihui Du
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, Jiangsu, People's Republic of China
| | - Zeyu Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, Jiangsu, People's Republic of China
| | - Cuie Guang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, Jiangsu, People's Republic of China
| | - Yingying Zhu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, Jiangsu, People's Republic of China
| | - Wanmeng Mu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, Jiangsu, People's Republic of China.
| |
Collapse
|
16
|
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
|
17
|
Welp A, Laser E, Seeger K, Haiß A, Hanke K, Faust K, Stichtenoth G, Fortmann-Grote C, Pagel J, Rupp J, Göpel W, Gembicki M, Scharf JL, Rody A, Herting E, Härtel C, Fortmann I. Effects of multistrain Bifidobacteria and Lactobacillus probiotics on HMO compositions after supplementation to pregnant women at threatening preterm delivery: design of the randomized clinical PROMO trial. Mol Cell Pediatr 2024; 11:6. [PMID: 39085734 PMCID: PMC11291828 DOI: 10.1186/s40348-024-00179-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND As an indigestible component of human breast milk, Human Milk Oligosaccharides (HMOs) play an important role as a substrate for the establishing microbiome of the newborn. They have further been shown to have beneficial effects on the immune system, lung and brain development. For preterm infants HMO composition of human breast milk may be of particular relevance since the establishment of a healthy microbiome is challenged by multiple disruptive factors associated with preterm birth, such as cesarean section, hospital environment and perinatal antibiotic exposure. In a previous study it has been proposed that maternal probiotic supplementation during late stages of pregnancy may change the HMO composition in human milk. However, there is currently no study on pregnancies which are threatened to preterm birth. Furthermore, HMO composition has not been investigated in association with clinically relevant outcomes of vulnerable infants including inflammation-mediated diseases such as sepsis, necrotizing enterocolitis (NEC) or chronic lung disease. MAIN BODY A randomized controlled intervention study (PROMO = probiotics for human milk oligosaccharides) has been designed to analyze changes in HMO composition of human breast milk after supplementation of probiotics (Lactobacillus acidophilus, Bifidobacterium lactis and Bifidobacterium infantis) in pregnancies at risk for preterm birth. The primary endpoint is HMO composition of 3-fucosyllactose and 3'-sialyllactose in expressed breast milk. We estimate that probiotic intervention will increase these two HMO levels by 50% according to the standardized mean difference between treatment and control groups. As secondary outcomes we will measure preterm infants' clinical outcomes (preterm birth, sepsis, weight gain growth, gastrointestinal complications) and effects on microbiome composition in the rectovaginal tract of mothers at delivery and in the gut of term and preterm infants by sequencing at high genomic resolution. Therefore, we will longitudinally collect bio samples in the first 4 weeks after birth as well as in follow-up investigations at 3 months, one year, and five years of age. CONCLUSIONS We estimate that probiotic intervention will increase these two HMO levels by 50% according to the standardized mean difference between treatment and control groups. The PROMO study will gain insight into the microbiome-HMO interaction at the fetomaternal interface and its consequences for duration of pregnancy and outcome of infants.
Collapse
Affiliation(s)
- A Welp
- Department of Gynecology and Obstetrics, University Hospital of Lübeck, Lübeck, Germany.
| | - E Laser
- Department of Pediatrics, University Hospital of Lübeck, Lübeck, Germany
| | - K Seeger
- Institute of Chemistry and Metabolomics, University of Lübeck, Lübeck, Germany
| | - A Haiß
- Department of Pediatrics, University Hospital of Lübeck, Lübeck, Germany
| | - K Hanke
- Department of Pediatrics, University Hospital of Lübeck, Lübeck, Germany
| | - K Faust
- Department of Pediatrics, University Hospital of Lübeck, Lübeck, Germany
| | - G Stichtenoth
- Department of Pediatrics, University Hospital of Lübeck, Lübeck, Germany
| | - C Fortmann-Grote
- Department of Microbial Population Biology, Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - J Pagel
- Department of Pediatrics, University Hospital of Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research, Lübeck, Germany
| | - J Rupp
- German Center for Infection Research, Lübeck, Germany
- Institute for Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - W Göpel
- Department of Pediatrics, University Hospital of Lübeck, Lübeck, Germany
| | - M Gembicki
- Department of Gynecology and Obstetrics, University Hospital of Lübeck, Lübeck, Germany
| | - J L Scharf
- Department of Gynecology and Obstetrics, University Hospital of Lübeck, Lübeck, Germany
| | - A Rody
- Department of Gynecology and Obstetrics, University Hospital of Lübeck, Lübeck, Germany
| | - E Herting
- Department of Pediatrics, University Hospital of Lübeck, Lübeck, Germany
| | - C Härtel
- Department of Pediatrics, University of Würzburg, Würzburg, Germany
| | - I Fortmann
- Department of Pediatrics, University Hospital of Lübeck, Lübeck, Germany
- German Center for Infection Research, Lübeck, Germany
| |
Collapse
|
18
|
Zhang L, Wu Z, Kang M, Wang J, Tan B. Utilization of Ningxiang pig milk oligosaccharides by Akkermansia muciniphila in vitro fermentation: enhancing neonatal piglet survival. Front Microbiol 2024; 15:1430276. [PMID: 38933035 PMCID: PMC11199860 DOI: 10.3389/fmicb.2024.1430276] [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: 05/09/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024] Open
Abstract
Akkermansia muciniphila (A. muciniphila), an intestinal symbiont residing in the mucosal layer, shows promise as a probiotic. Our previous study found that the abundance of A. muciniphila was significantly higher in Ningxiang suckling piglets compared to other breeds, suggesting that early breast milk may play a crucial role. This study examines A. muciniphila's ability to utilize Ningxiang pig milk oligosaccharides. We discovered that A. muciniphila can thrive on both Ningxiang pig colostrum and purified pig milk oligosaccharides. Genetic analysis has shown that A. muciniphila harbors essential glycan-degrading enzymes, enabling it to effectively break down a broad spectrum of oligosaccharides. Our findings demonstrate that A. muciniphila can degrade pig milk oligosaccharides structures such as 3'-FL, 3'-SL, LNT, and LNnT, producing short-chain fatty acids in the process. The hydrolysis of these host-derived glycan structures enhances A. muciniphila's symbiotic interactions with other beneficial gut bacteria, contributing to a dynamic microbial ecological network. The capability of A. muciniphila to utilize pig milk oligosaccharides allows it to establish itself in the intestines of newborn piglets, effectively colonizing the mucosal layer early in life. This early colonization is key in supporting both mucosal and metabolic health, which is critical for enhancing piglet survival during lactation.
Collapse
Affiliation(s)
- Longlin Zhang
- Key Laboratory for Quality Regulation of Livestock and Poultry Products of Hunan Province, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Yuelushan Laboratory, Changsha, China
| | - Zichen Wu
- Key Laboratory for Quality Regulation of Livestock and Poultry Products of Hunan Province, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Yuelushan Laboratory, Changsha, China
| | - Meng Kang
- Key Laboratory for Quality Regulation of Livestock and Poultry Products of Hunan Province, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Yuelushan Laboratory, Changsha, China
| | - Jing Wang
- Key Laboratory for Quality Regulation of Livestock and Poultry Products of Hunan Province, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Yuelushan Laboratory, Changsha, China
| | - Bie Tan
- Key Laboratory for Quality Regulation of Livestock and Poultry Products of Hunan Province, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Yuelushan Laboratory, Changsha, China
| |
Collapse
|
19
|
Lu B, Liao SM, Liang SJ, Li JX, Liu XH, Huang RB, Zhou GP. NMR Studies of the Interactions between Sialyllactoses and the Polysialytransferase Domain for Polysialylation Inhibition. Curr Issues Mol Biol 2024; 46:5682-5700. [PMID: 38921011 PMCID: PMC11201969 DOI: 10.3390/cimb46060340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 04/30/2024] [Accepted: 05/26/2024] [Indexed: 06/27/2024] Open
Abstract
It is known that sialyllactose (SL) in mammalians is a major source of sialic acid (Sia), which can further form cytidine monophosphate sialic acid (CMP-Sia), and the final product is polysialic acid (polySia) using polysialyltransferases (polySTs) on the neural cell adhesion molecule (NCAM). This process is called NCAM polysialylation. The overexpression of polysialylation is strongly related to cancer cell migration, invasion, and metastasis. In order to inhibit the overexpression of polysialylation, in this study, SL was selected as an inhibitor to test whether polysialylation could be inhibited. Our results suggest that the interactions between the polysialyltransferase domain (PSTD) in polyST and CMP-Siaand the PSTD and polySia could be inhibited when the 3'-sialyllactose (3'-SL) or 6'-sialyllactose (6'-SL) concentration is about 0.5 mM or 6'-SL and 3 mM, respectively. The results also show that SLs (particularly for 3'-SL) are the ideal inhibitors compared with another two inhibitors, low-molecular-weight heparin (LMWH) and cytidine monophosphate (CMP), because 3'-SL can not only be used to inhibit NCAM polysialylation, but is also one of the best supplements for infant formula and the gut health system.
Collapse
Affiliation(s)
- Bo Lu
- National Engineering Research Center for Non-Food Biorefinery, Guangxi Academy of Sciences, 98 Daling Road, Nanning 530007, China; (B.L.); (S.-M.L.); (S.-J.L.); (J.-X.L.)
| | - Si-Ming Liao
- National Engineering Research Center for Non-Food Biorefinery, Guangxi Academy of Sciences, 98 Daling Road, Nanning 530007, China; (B.L.); (S.-M.L.); (S.-J.L.); (J.-X.L.)
| | - Shi-Jie Liang
- National Engineering Research Center for Non-Food Biorefinery, Guangxi Academy of Sciences, 98 Daling Road, Nanning 530007, China; (B.L.); (S.-M.L.); (S.-J.L.); (J.-X.L.)
| | - Jian-Xiu Li
- National Engineering Research Center for Non-Food Biorefinery, Guangxi Academy of Sciences, 98 Daling Road, Nanning 530007, China; (B.L.); (S.-M.L.); (S.-J.L.); (J.-X.L.)
| | - Xue-Hui Liu
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China;
| | - Ri-Bo Huang
- National Engineering Research Center for Non-Food Biorefinery, Guangxi Academy of Sciences, 98 Daling Road, Nanning 530007, China; (B.L.); (S.-M.L.); (S.-J.L.); (J.-X.L.)
- Life Science and Technology College, Guangxi University, Nanning 530004, China
- Rocky Mount Life Science Institute, Rocky Mount, NC 27804, USA
| | - Guo-Ping Zhou
- National Engineering Research Center for Non-Food Biorefinery, Guangxi Academy of Sciences, 98 Daling Road, Nanning 530007, China; (B.L.); (S.-M.L.); (S.-J.L.); (J.-X.L.)
- Rocky Mount Life Science Institute, Rocky Mount, NC 27804, USA
| |
Collapse
|
20
|
Nishihara K, Villot C, Cangiano L, Guan LL, Steele M. Bacteria colonization and gene expression related to immune function in colon mucosa is associated with growth in neonatal calves regardless of live yeast supplementation. J Anim Sci Biotechnol 2024; 15:76. [PMID: 38835065 DOI: 10.1186/s40104-024-01030-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/01/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND As Holstein calves are susceptible to gastrointestinal disorders during the first week of life, understanding how intestinal immune function develops in neonatal calves is important to promote better intestinal health. Feeding probiotics in early life may contribute to host intestinal health by facilitating beneficial bacteria colonization and developing intestinal immune function. The objective of this study was to characterize the impact of early life yeast supplementation and growth on colon mucosa-attached bacteria and host immune function. RESULTS Twenty Holstein bull calves received no supplementation (CON) or Saccharomyces cerevisiae boulardii (SCB) from birth to 5 d of life. Colon tissue biopsies were taken within 2 h of life (D0) before the first colostrum feeding and 3 h after the morning feeding at d 5 of age (D5) to analyze mucosa-attached bacteria and colon transcriptome. Metagenome sequencing showed that there was no difference in α and β diversity of mucosa-attached bacteria between day and treatment, but bacteria related to diarrhea were more abundant in the colon mucosa on D0 compared to D5. In addition, qPCR indicated that the absolute abundance of Escherichia coli (E. coli) decreased in the colon mucosa on D5 compared to D0; however, that of Bifidobacterium, Lactobacillus, and Faecalibacterium prausnitzii, which could competitively exclude E. coli, increased in the colon mucosa on D5 compared to D0. RNA-sequencing showed that there were no differentially expressed genes between CON and SCB, but suggested that pathways related to viral infection such as "Interferon Signaling" were activated in the colon mucosa of D5 compared to D0. CONCLUSIONS Growth affected mucosa-attached bacteria and host immune function in the colon mucosa during the first 5 d of life in dairy calves independently of SCB supplementation. During early life, opportunistic pathogens may decrease due to intestinal environmental changes by beneficial bacteria and/or host immune function. Predicted activation of immune function-related pathways may be the result of host immune function development or suggest other antigens in the intestine during early life. Further studies focusing on the other antigens and host immune function in the colon mucosa are required to better understand intestinal immune function development.
Collapse
Affiliation(s)
- Koki Nishihara
- Department of Animal Biosciences, Animal Science and Nutrition, University of Guelph, Guelph, ON, N1G 1Y2, Canada
| | - Clothilde Villot
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, T6G 2P5, Canada
- Lallemand SAS, Blagnac, F-31702, France
| | - Lautaro Cangiano
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Le Luo Guan
- Faculty of Land and Food Systems, The University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Michael Steele
- Department of Animal Biosciences, Animal Science and Nutrition, University of Guelph, Guelph, ON, N1G 1Y2, Canada.
- Present Address: Department of Animal Biosciences, University of Guelph, Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
21
|
Mulinge MM, Abisi HK, Kabahweza HM, Okutoyi L, Wamalwa DC, Nduati RW. The Role of Maternal Secretor Status and Human Milk Oligosaccharides on Early Childhood Development: A Systematic Review and Meta-Analysis. Breastfeed Med 2024; 19:409-424. [PMID: 38577928 DOI: 10.1089/bfm.2023.0274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
Background: Breast milk is the gold standard of infant nutrition, delivering nutrients and bioactive molecules as needed to support optimal infant growth and cognitive development. Increasing evidence links human milk oligosaccharides (HMOs) to these early childhood development milestones. Aims: To summarize and synthesize the evidence relating to HMOs and infant brain development, physical growth, and cognitive development. In addition, HMO concentrations in secretor and nonsecretor mothers were compared via a meta-analysis. Study Design: A systematic review and meta-analysis were carried out in accordance with the PRISMA statement. This review used three databases (PubMed, Scopus, and Web of Science) and was limited to English-language articles published between 2000 and June 30, 2023. Results: The initial searches yielded 245 articles, 27 of which were included in the systematic review and 12 in the meta-analysis. The meta-analysis revealed a substantial between-study heterogeneity, I2 = 97.3%. The pooled effect was 0.21 (95% CI: -0.41 to 0.83; p = 0.484), indicating that secretors had higher HMO concentrations, although this difference was not statistically significant. At one month of age, 2'FL, 3FL, and 3'SL play an important role in brain maturation and thus play a critical role in cognitive development. Secretors produce higher concentrations of 2'FL and 3'SL, explaining the benefits to infants of secretor mothers. Growth velocity was correlated to fucosylated and sialylated HMO concentrations, with lower concentrations linked to stunting. Conclusions: According to evidence from the systematically reviewed articles, HMOs are essential for a child's early development, but the extent to which they have an impact depends on maternal secretor status.
Collapse
Affiliation(s)
- Martin M Mulinge
- Department of Biochemistry, School of Medicine, University of Nairobi, Nairobi, Kenya
| | - Hellen K Abisi
- Department of Biochemistry, School of Medicine, University of Nairobi, Nairobi, Kenya
| | - Hellen M Kabahweza
- Department of Pediatric Hematology & Oncology, Joint Clinical Research Centre, Kampala, Uganda
| | - Lydia Okutoyi
- Department of Health Care Quality, Kenyatta National Hospital, Nairobi, Kenya
| | - Dalton C Wamalwa
- Department of Paediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Ruth W Nduati
- Department of Paediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| |
Collapse
|
22
|
Endika MF, Barnett DJM, Klostermann CE, Kok N, Schols HA, Nauta A, Arts ICW, Penders J, Venema K, Smidt H. Seeding and feeding milestones: the role of human milk microbes and oligosaccharides in the temporal development of infant gut microbiota. GUT MICROBIOME (CAMBRIDGE, ENGLAND) 2024; 5:e7. [PMID: 39776540 PMCID: PMC11706684 DOI: 10.1017/gmb.2024.5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 04/24/2024] [Accepted: 04/26/2024] [Indexed: 01/11/2025]
Abstract
Breastfeeding represents a strong selective factor for shaping the infant gut microbiota. Besides providing nutritional requirements for the infant, human milk is a key source of oligosaccharides, human milk oligosaccharides (HMOs), and diverse microbes in early life. This study aimed to evaluate the influence of human milk microbiota and oligosaccharides on the composition of infant faecal microbiota at one, three, and nine months postpartum. We profiled milk microbiota, HMOs, and infant faecal microbiota from 23 mother-infant pairs at these time points. The predominant genera in milk samples were Streptococcus, Staphylococcus, and an unclassified Enterobacteriaceae genus-level taxon (Enterobacteriaceae uncl.), whereas the infant faecal microbiota was predominated by Bifidobacterium, Bacteroides, and Enterobacteriaceae uncl. Mother-infant dyads frequently shared bacterial amplicon sequence variants (ASVs) belonging to the genera Bifidobacterium, Streptococcus, Enterobacteriaceae uncl., Veillonella, Bacteroides, and Haemophilus. The individual HMO concentrations in the milk showed either no change or decreased over the lactation period, except for 3-fucosyllactose (3-FL), which increased. Neither maternal secretor status nor HMO concentrations were significantly associated with microbiota composition at the different ages or the bacterial ASVs of maternal milk and infant faeces. This study suggests an age-dependent role of milk microbes in shaping the gut microbiota, while variations in HMO concentrations show limited influence.
Collapse
Affiliation(s)
- Martha F. Endika
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | - David J. M. Barnett
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, The Netherlands
- Department of Medical Microbiology, Infectious Diseases and Infection Prevention, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Cynthia E. Klostermann
- Biobased Chemistry and Technology, Wageningen University & Research, Wageningen, The Netherlands
- Laboratory of Food Chemistry, Wageningen University & Research, Wageningen, The Netherlands
| | - Noortje Kok
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Henk A. Schols
- Laboratory of Food Chemistry, Wageningen University & Research, Wageningen, The Netherlands
| | - Arjen Nauta
- FrieslandCampina, Amersfoort, The Netherlands
| | - Ilja C. W. Arts
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, The Netherlands
| | - John Penders
- Department of Medical Microbiology, Infectious Diseases and Infection Prevention, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Koen Venema
- Centre for Healthy Eating & Food Innovation (HEFI), Maastricht University – campus Venlo, Venlo, The Netherlands
| | - Hauke Smidt
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| |
Collapse
|
23
|
Moya-Gonzálvez EM, Zeuner B, Thorhallsson AT, Holck J, Palomino-Schätzlein M, Rodríguez-Díaz J, Meyer AS, Yebra MJ. Synthesis of fucosyllactose using α-L-fucosidases GH29 from infant gut microbial metagenome. Appl Microbiol Biotechnol 2024; 108:338. [PMID: 38771321 PMCID: PMC11108932 DOI: 10.1007/s00253-024-13178-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/23/2024] [Accepted: 05/10/2024] [Indexed: 05/22/2024]
Abstract
Fucosyl-oligosaccharides (FUS) provide many health benefits to breastfed infants, but they are almost completely absent from bovine milk, which is the basis of infant formula. Therefore, there is a growing interest in the development of enzymatic transfucosylation strategies for the production of FUS. In this work, the α-L-fucosidases Fuc2358 and Fuc5372, previously isolated from the intestinal bacterial metagenome of breastfed infants, were used to synthesize fucosyllactose (FL) by transfucosylation reactions using p-nitrophenyl-α-L-fucopyranoside (pNP-Fuc) as donor and lactose as acceptor. Fuc2358 efficiently synthesized the major fucosylated human milk oligosaccharide (HMO) 2'-fucosyllactose (2'FL) with a 35% yield. Fuc2358 also produced the non-HMO FL isomer 3'-fucosyllactose (3'FL) and traces of non-reducing 1-fucosyllactose (1FL). Fuc5372 showed a lower transfucosylation activity compared to Fuc2358, producing several FL isomers, including 2'FL, 3'FL, and 1FL, with a higher proportion of 3'FL. Site-directed mutagenesis using rational design was performed to increase FUS yields in both α-L-fucosidases, based on structural models and sequence identity analysis. Mutants Fuc2358-F184H, Fuc2358-K286R, and Fuc5372-R230K showed a significantly higher ratio between 2'FL yields and hydrolyzed pNP-Fuc than their respective wild-type enzymes after 4 h of transfucosylation. The results with the Fuc2358-F184W and Fuc5372-W151F mutants showed that the residues F184 of Fuc2358 and W151 of Fuc5372 could have an effect on transfucosylation regioselectivity. Interestingly, phenylalanine increases the selectivity for α-1,2 linkages and tryptophan for α-1,3 linkages. These results give insight into the functionality of the active site amino acids in the transfucosylation activity of the GH29 α-L-fucosidases Fuc2358 and Fuc5372. KEY POINTS: Two α-L-fucosidases from infant gut bacterial microbiomes can fucosylate glycans Transfucosylation efficacy improved by tailored point-mutations in the active site F184 of Fuc2358 and W151 of Fuc5372 seem to steer transglycosylation regioselectivity.
Collapse
Affiliation(s)
- Eva M Moya-Gonzálvez
- Laboratorio de Bacterias Lácticas y Probióticos, Departamento de Biotecnología de Alimentos, Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Valencia, Spain
| | - Birgitte Zeuner
- Protein Chemistry and Enzyme Technology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Albert Th Thorhallsson
- Protein Chemistry and Enzyme Technology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Jesper Holck
- Protein Chemistry and Enzyme Technology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | | | - Jesús Rodríguez-Díaz
- Departamento de Microbiología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Anne S Meyer
- Protein Chemistry and Enzyme Technology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - María J Yebra
- Laboratorio de Bacterias Lácticas y Probióticos, Departamento de Biotecnología de Alimentos, Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Valencia, Spain.
| |
Collapse
|
24
|
Yao Q, Gao Y, Zheng N, Delcenserie V, Wang J. Unlocking the mysteries of milk oligosaccharides: Structure, metabolism, and function. Carbohydr Polym 2024; 332:121911. [PMID: 38431414 DOI: 10.1016/j.carbpol.2024.121911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/18/2024] [Accepted: 01/31/2024] [Indexed: 03/05/2024]
Abstract
Milk oligosaccharides (MOs), complex carbohydrates prevalent in human breast milk, play a vital role in infant nutrition. Serving as prebiotics, they inhibit pathogen adherence, modulate the immune system, and support newborn brain development. Notably, MOs demonstrate significant variations in concentration and composition, both across different species and within the same species. These characteristics of MOs lead to several compelling questions: (i) What distinct beneficial functions do MOs offer and how do the functions vary along with their structural differences? (ii) In what ways do MOs in human milk differ from those in other mammals, and what factors drive these unique profiles? (iii) What are the emerging applications of MOs, particularly in the context of their incorporation into infant formula? This review delves into the structural characteristics, quantification methods, and species-specific concentration differences of MOs. It highlights the critical role of human MOs in infant growth and their potential applications, providing substantial evidence to enhance infant health and development.
Collapse
Affiliation(s)
- Qianqian Yao
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China; Department of Food Science, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Yanan Gao
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Nan Zheng
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Veronique Delcenserie
- Department of Food Science, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Jiaqi Wang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China.
| |
Collapse
|
25
|
Shaw C, Weimer BC, Gann R, Desai PT, Shah JD. The Yin and Yang of pathogens and probiotics: interplay between Salmonella enterica sv. Typhimurium and Bifidobacterium infantis during co-infection. Front Microbiol 2024; 15:1387498. [PMID: 38812689 PMCID: PMC11133690 DOI: 10.3389/fmicb.2024.1387498] [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: 02/17/2024] [Accepted: 04/12/2024] [Indexed: 05/31/2024] Open
Abstract
Probiotic bacteria have been proposed as an alternative to antibiotics for the control of antimicrobial resistant enteric pathogens. The mechanistic details of this approach remain unclear, in part because pathogen reduction appears to be both strain and ecology dependent. Here we tested the ability of five probiotic strains, including some from common probiotic genera Lactobacillus and Bifidobacterium, to reduce binding of Salmonella enterica sv. Typhimurium to epithelial cells in vitro. Bifidobacterium longum subsp. infantis emerged as a promising strain; however, S. Typhimurium infection outcome in epithelial cells was dependent on inoculation order, with B. infantis unable to rescue host cells from preceding or concurrent infection. We further investigated the complex mechanisms underlying this interaction between B. infantis, S. Typhimurium, and epithelial cells using a multi-omics approach that included gene expression and altered metabolism via metabolomics. Incubation with B. infantis repressed apoptotic pathways and induced anti-inflammatory cascades in epithelial cells. In contrast, co-incubation with B. infantis increased in S. Typhimurium the expression of virulence factors, induced anaerobic metabolism, and repressed components of arginine metabolism as well as altering the metabolic profile. Concurrent application of the probiotic and pathogen notably generated metabolic profiles more similar to that of the probiotic alone than to the pathogen, indicating a central role for metabolism in modulating probiotic-pathogen-host interactions. Together these data imply crosstalk via small molecules between the epithelial cells, pathogen and probiotic that consistently demonstrated unique molecular mechanisms specific probiotic/pathogen the individual associations.
Collapse
Affiliation(s)
| | - Bart C. Weimer
- Department of Population Health and Reproduction, School of Veterinary Medicine, 100K Pathogen Genome Project, University of California, Davis, Davis, CA, United States
| | | | | | | |
Collapse
|
26
|
Pitt J, Bond J, Roper J, Tenning P, Mukherjea R, Evans K, Saarinen MT, Anglenius H, Hirvonen J, Hasselwander O, Lim A. A 21-day safety evaluation of biotechnologically produced 3-fucosyllactose (3-FL) in neonatal farm piglets to support use in infant formulas. Food Chem Toxicol 2024; 187:114592. [PMID: 38493976 DOI: 10.1016/j.fct.2024.114592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/07/2024] [Accepted: 03/09/2024] [Indexed: 03/19/2024]
Abstract
3-Fucosyllactose (3-FL) is one of the most abundant fucosylated oligosaccharides in human breast milk and is an approved infant formula ingredient world-wide. 3-FL functions as a prebiotic to promote early microbial colonization of the gut, increase pathogen resistance and modulate immune responses. To investigate safety and potential gut microbiota effects, 3-FL was fed for 21-days to farm piglets beginning on Postnatal Day (PND) 2. Fructooligosaccharide (FOS), an approved infant formula ingredient, was used as a reference control. Standard toxicological endpoints were evaluated, and the gut microbiota were assessed. Neither 3-FL (245.77 and 489.72 mg/kg/day for males and 246.57 and 494.18 mg/kg/day for females) nor FOS (489.44 and 496.33 mg/kg/day males and females, respectively) produced any adverse differences in growth, food intake or efficiency, clinical observations, or clinical or anatomic pathology changes. Differences in the gut microbiota after 3-FL consumption (versus control and FOS groups) included the absence of Bifidobacterium species from the piglets, enrichment of Prevotellamassilia timonensis, Blautia species, Mediterranea massiliensis, Lachnospiraceae incertae sedis, and Eubacterium coprostanoligens and lower relative abundance of Allisonella histaminiformans and Roseburia inulinivorans. This study further supports the safe use of 3-FL produced using biotechnology as a nutritional ingredient in foods.
Collapse
Affiliation(s)
- Jeffrey Pitt
- International Flavors & Fragrances, Larkin Laboratory, 1803 Larkin Center Drive, Midland, MI, 48642, USA.
| | - Jennifer Bond
- Charles River (CR-MWN), 54943 N. Main Street, Mattawan, MI, 49071, USA; Labcorp Drug Development, 671 South Meridian Road, Greenfield, IN, 46140, USA
| | - Jason Roper
- DuPont Stine-Haskell, 1090 Elkton Rd, Newark, DE, 19714, USA; Teva Pharmaceuticals, 145 Brandywine Parkway, West Chester, PA, 19380, USA
| | - Paul Tenning
- International Flavors & Fragrances, Leiden Bio Science Park, Galileiweg 8, 2333 BD, Leiden, the Netherlands
| | - Ratna Mukherjea
- DuPont Stine-Haskell, 1090 Elkton Rd, Newark, DE, 19714, USA; Benson Hill, 1001 N Warson Rd, St. Louis, MO, 63132, USA
| | - Kara Evans
- International Flavors & Fragrances, 3329 Agriculture Drive, Madison, WI, 53716, USA
| | - Markku T Saarinen
- International Flavors & Fragrances, Health & Biosciences Danisco Sweeteners Oy, Sokeritehtaantie 20, 02460, Kantvik, Finland
| | - Heli Anglenius
- International Flavors & Fragrances, Health & Biosciences Danisco Sweeteners Oy, Sokeritehtaantie 20, 02460, Kantvik, Finland
| | - Johanna Hirvonen
- International Flavors & Fragrances, Health & Biosciences Danisco Sweeteners Oy, Sokeritehtaantie 20, 02460, Kantvik, Finland
| | - Oliver Hasselwander
- International Flavors & Fragrances, Health & Biosciences, c/o Danisco UK Ltd., Reigate, RH2 9PW, United Kingdom
| | - Angela Lim
- International Flavors & Fragrances, DuPont Experimental Station, Bldg. 353, 200 Powder Mill Rd, Wilmington, DE, 19803, USA
| |
Collapse
|
27
|
Hilliard MA, Sela DA. Transmission and Persistence of Infant Gut-Associated Bifidobacteria. Microorganisms 2024; 12:879. [PMID: 38792709 PMCID: PMC11124121 DOI: 10.3390/microorganisms12050879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/18/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
Bifidobacterium infantis are the primary colonizers of the infant gut, yet scientific research addressing the transmission of the genus Bifidobacterium to infants remains incomplete. This review examines microbial reservoirs of infant-type Bifidobacterium that potentially contribute to infant gut colonization. Accordingly, strain inheritance from mother to infant via the fecal-oral route is likely contingent on the bifidobacterial strain and phenotype, whereas transmission via the vaginal microbiota may be restricted to Bifidobacterium breve. Additional reservoirs include breastmilk, horizontal transfer from the environment, and potentially in utero transfer. Given that diet is a strong predictor of Bifidobacterium colonization in early life and the absence of Bifidobacterium is observed regardless of breastfeeding, it is likely that additional factors are responsible for bifidobacterial colonization early in life.
Collapse
Affiliation(s)
- Margaret A. Hilliard
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA;
- Organismic and Evolutionary Biology Graduate Program, University of Massachusetts, Amherst, MA 01003, USA
| | - David A. Sela
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA;
- Organismic and Evolutionary Biology Graduate Program, University of Massachusetts, Amherst, MA 01003, USA
- Department of Nutrition, University of Massachusetts, Amherst, MA 01003, USA
- Department of Microbiology, University of Massachusetts, Amherst, MA 01003, USA
- Department of Microbiology & Physiological Systems and Center for Microbiome Research, University of Massachusetts Medical School, Worcester, MA 01605, USA
| |
Collapse
|
28
|
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
|
29
|
Dedola S, Ahmadipour S, de Andrade P, Baker AN, Boshra AN, Chessa S, Gibson MI, Hernando PJ, Ivanova IM, Lloyd JE, Marín MJ, Munro-Clark AJ, Pergolizzi G, Richards SJ, Ttofi I, Wagstaff BA, Field RA. Sialic acids in infection and their potential use in detection and protection against pathogens. RSC Chem Biol 2024; 5:167-188. [PMID: 38456038 PMCID: PMC10915975 DOI: 10.1039/d3cb00155e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 12/12/2023] [Indexed: 03/09/2024] Open
Abstract
In structural terms, the sialic acids are a large family of nine carbon sugars based around an alpha-keto acid core. They are widely spread in nature, where they are often found to be involved in molecular recognition processes, including in development, immunology, health and disease. The prominence of sialic acids in infection is a result of their exposure at the non-reducing terminus of glycans in diverse glycolipids and glycoproteins. Herein, we survey representative aspects of sialic acid structure, recognition and exploitation in relation to infectious diseases, their diagnosis and prevention or treatment. Examples covered span influenza virus and Covid-19, Leishmania and Trypanosoma, algal viruses, Campylobacter, Streptococci and Helicobacter, and commensal Ruminococci.
Collapse
Affiliation(s)
- Simone Dedola
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
- Iceni Glycoscience Ltd, Norwich Research Park Norwich NR4 7TJ UK
| | - Sanaz Ahmadipour
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
| | - Peterson de Andrade
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
| | - Alexander N Baker
- Department of Chemistry, University of Warwick Gibbet Hill Road Coventry CV4 7AL UK
| | - Andrew N Boshra
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Assiut University Assiut 71526 Egypt
| | - Simona Chessa
- Iceni Glycoscience Ltd, Norwich Research Park Norwich NR4 7TJ UK
| | - Matthew I Gibson
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
- Department of Chemistry, University of Warwick Gibbet Hill Road Coventry CV4 7AL UK
- Division of Biomedical Sciences, Warwick Medical School Coventry CV4 7AL UK
| | - Pedro J Hernando
- Iceni Glycoscience Ltd, Norwich Research Park Norwich NR4 7TJ UK
| | - Irina M Ivanova
- Iceni Glycoscience Ltd, Norwich Research Park Norwich NR4 7TJ UK
| | - Jessica E Lloyd
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
| | - María J Marín
- School of Chemistry, University of East Anglia, Norwich Research Park Norwich NR4 7TJ UK
| | - Alexandra J Munro-Clark
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
| | | | - Sarah-Jane Richards
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
- Department of Chemistry, University of Warwick Gibbet Hill Road Coventry CV4 7AL UK
| | - Iakovia Ttofi
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
- Iceni Glycoscience Ltd, Norwich Research Park Norwich NR4 7TJ UK
| | - Ben A Wagstaff
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
| | - Robert A Field
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
- Iceni Glycoscience Ltd, Norwich Research Park Norwich NR4 7TJ UK
| |
Collapse
|
30
|
Mercer EM, Ramay HR, Moossavi S, Laforest-Lapointe I, Reyna ME, Becker AB, Simons E, Mandhane PJ, Turvey SE, Moraes TJ, Sears MR, Subbarao P, Azad MB, Arrieta MC. Divergent maturational patterns of the infant bacterial and fungal gut microbiome in the first year of life are associated with inter-kingdom community dynamics and infant nutrition. MICROBIOME 2024; 12:22. [PMID: 38326891 PMCID: PMC10848358 DOI: 10.1186/s40168-023-01735-3] [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: 08/27/2023] [Accepted: 12/01/2023] [Indexed: 02/09/2024]
Abstract
BACKGROUND The gut microbiome undergoes primary ecological succession over the course of early life before achieving ecosystem stability around 3 years of age. These maturational patterns have been well-characterized for bacteria, but limited descriptions exist for other microbiota members, such as fungi. Further, our current understanding of the prevalence of different patterns of bacterial and fungal microbiome maturation and how inter-kingdom dynamics influence early-life microbiome establishment is limited. RESULTS We examined individual shifts in bacterial and fungal alpha diversity from 3 to 12 months of age in 100 infants from the CHILD Cohort Study. We identified divergent patterns of gut bacterial or fungal microbiome maturation in over 40% of infants, which were characterized by differences in community composition, inter-kingdom dynamics, and microbe-derived metabolites in urine, suggestive of alterations in the timing of ecosystem transitions. Known microbiome-modifying factors, such as formula feeding and delivery by C-section, were associated with atypical bacterial, but not fungal, microbiome maturation patterns. Instead, fungal microbiome maturation was influenced by prenatal exposure to artificially sweetened beverages and the bacterial microbiome, emphasizing the importance of inter-kingdom dynamics in early-life colonization patterns. CONCLUSIONS These findings highlight the ecological and environmental factors underlying atypical patterns of microbiome maturation in infants, and the need to incorporate multi-kingdom and individual-level perspectives in microbiome research to improve our understandings of gut microbiome maturation patterns in early life and how they relate to host health. Video Abstract.
Collapse
Affiliation(s)
- Emily M Mercer
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- International Microbiome Center, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute (ACHRI), Calgary, AB, Canada
| | - Hena R Ramay
- International Microbiome Center, University of Calgary, Calgary, AB, Canada
| | - Shirin Moossavi
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Louvain, Belgium
- VIB Center for Microbiology, VIB, Louvain, Belgium
| | | | - Myrtha E Reyna
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
- Department of Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Allan B Becker
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| | - Elinor Simons
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| | - Piush J Mandhane
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Stuart E Turvey
- Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Theo J Moraes
- Department of Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Malcolm R Sears
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Padmaja Subbarao
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
- Department of Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Meghan B Azad
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
- Manitoba Interdisciplinary Lactation Centre (MILC), Winnipeg, MB, Canada
| | - Marie-Claire Arrieta
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada.
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada.
- International Microbiome Center, University of Calgary, Calgary, AB, Canada.
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada.
- Alberta Children's Hospital Research Institute (ACHRI), Calgary, AB, Canada.
| |
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
|
McDonald AG, Lisacek F. Simulated digestions of free oligosaccharides and mucin-type O-glycans reveal a potential role for Clostridium perfringens. Sci Rep 2024; 14:1649. [PMID: 38238389 PMCID: PMC10796942 DOI: 10.1038/s41598-023-51012-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 12/29/2023] [Indexed: 01/22/2024] Open
Abstract
The development of a stable human gut microbiota occurs within the first year of life. Many open questions remain about how microfloral species are influenced by the composition of milk, in particular its content of human milk oligosaccharides (HMOs). The objective is to investigate the effect of the human HMO glycome on bacterial symbiosis and competition, based on the glycoside hydrolase (GH) enzyme activities known to be present in microbial species. We extracted from UniProt a list of all bacterial species catalysing glycoside hydrolase activities (EC 3.2.1.-), cross-referencing with the BRENDA database, and obtained a set of taxonomic lineages and CAZy family data. A set of 13 documented enzyme activities was selected and modelled within an enzyme simulator according to a method described previously in the context of biosynthesis. A diverse population of experimentally observed HMOs was fed to the simulator, and the enzymes matching specific bacterial species were recorded, based on their appearance of individual enzymes in the UniProt dataset. Pairs of bacterial species were identified that possessed complementary enzyme profiles enabling the digestion of the HMO glycome, from which potential symbioses could be inferred. Conversely, bacterial species having similar GH enzyme profiles were considered likely to be in competition for the same set of dietary HMOs within the gut of the newborn. We generated a set of putative biodegradative networks from the simulator output, which provides a visualisation of the ability of organisms to digest HMO and mucin-type O-glycans. B. bifidum, B. longum and C. perfringens species were predicted to have the most diverse GH activity and therefore to excel in their ability to digest these substrates. The expected cooperative role of Bifidobacteriales contrasts with the surprising capacities of the pathogen. These findings indicate that potential pathogens may associate in human gut based on their shared glycoside hydrolase digestive apparatus, and which, in the event of colonisation, might result in dysbiosis. The methods described can readily be adapted to other enzyme categories and species as well as being easily fine-tuneable if new degrading enzymes are identified and require inclusion in the model.
Collapse
Affiliation(s)
- Andrew G McDonald
- Proteome Informatics Group, SIB Swiss Institute of Bioinformatics, 1211, Geneva, Switzerland.
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin 2, Ireland.
| | - Frédérique Lisacek
- Proteome Informatics Group, SIB Swiss Institute of Bioinformatics, 1211, Geneva, Switzerland.
- Computer Science Department, University of Geneva, Geneva, Switzerland.
- Section of Biology, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
33
|
Ioannou A, Berkhout MD, Scott WT, Blijenberg B, Boeren S, Mank M, Knol J, Belzer C. Resource sharing of an infant gut microbiota synthetic community in combinations of human milk oligosaccharides. THE ISME JOURNAL 2024; 18:wrae209. [PMID: 39423288 PMCID: PMC11542058 DOI: 10.1093/ismejo/wrae209] [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: 03/18/2024] [Revised: 06/18/2024] [Accepted: 10/17/2024] [Indexed: 10/21/2024]
Abstract
Quickly after birth, the gut microbiota is shaped via species acquisition and resource pressure. Breastmilk, and more specifically, human milk oligosaccharides are a determining factor in the formation of microbial communities and the interactions between bacteria. Prominent human milk oligosaccharide degraders have been rigorously characterized, but it is not known how the gut microbiota is shaped as a complex community. Here, we designed BIG-Syc, a synthetic community of 13 strains from the gut of vaginally born, breastfed infants. BIG-Syc replicated key compositional, metabolic, and proteomic characteristics of the gut microbiota of infants. Upon fermentation of a four and five human milk oligosaccharide mix, BIG-Syc demonstrated different compositional and proteomic profiles, with Bifidobacterium infantis and Bifidobacterium bifidum suppressing one another. The mix of five human milk oligosaccharides resulted in a more diverse composition with dominance of B. bifidum, whereas that with four human milk oligosaccharides supported the dominance of B. infantis, in four of six replicates. Reintroduction of bifidobacteria to BIG-Syc led to their engraftment and establishment of their niche. Based on proteomics and genome-scale metabolic models, we reconstructed the carbon source utilization and metabolite and gas production per strain. BIG-Syc demonstrated teamwork as cross-feeders utilized simpler carbohydrates, organic acids, and gases released from human milk oligosaccharide degraders. Collectively, our results showed that human milk oligosaccharides prompt resource-sharing for their complete degradation while leading to a different compositional and functional profile in the community. At the same time, BIG-Syc proved to be an accurate model for the representation of intra-microbe interactions.
Collapse
Affiliation(s)
- Athanasia Ioannou
- Laboratory of Microbiology, Wageningen University & Research, Stippeneng 4, Wageningen 6708WE, the Netherlands
| | - Maryse D Berkhout
- Laboratory of Microbiology, Wageningen University & Research, Stippeneng 4, Wageningen 6708WE, the Netherlands
| | - William T Scott
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, Stippeneng 4, Wageningen 6708WE, the Netherlands
- UNLOCK, Wageningen University & Research and Delft University of Technology, Stippeneng 2, Wageningen 6708WE, the Netherlands
| | | | - Sjef Boeren
- Laboratory of Biochemistry, Wageningen University & Research, Stippeneng 4, Wageningen 6708WE, the Netherlands
| | - Marko Mank
- Danone Nutricia Research, Uppsalalaan 12, Utrecht 3584CT, the Netherlands
| | - Jan Knol
- Danone Nutricia Research, Uppsalalaan 12, Utrecht 3584CT, the Netherlands
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University & Research, Stippeneng 4, Wageningen 6708WE, the Netherlands
| |
Collapse
|
34
|
Reens AL, Cosetta CM, Saur R, Trofimuk O, Brooker SL, Lee ML, Sun AK, McKenzie GJ, Button JE. Tunable control of B. infantis abundance and gut metabolites by co-administration of human milk oligosaccharides. Gut Microbes 2024; 16:2304160. [PMID: 38235736 PMCID: PMC10798361 DOI: 10.1080/19490976.2024.2304160] [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: 09/19/2023] [Accepted: 01/08/2024] [Indexed: 01/19/2024] Open
Abstract
Precision engineering of the gut microbiome holds promise as an effective therapeutic approach for diseases associated with a disruption in this microbial community. Engrafting a live biotherapeutic product (LBP) in a predictable, controllable manner is key to the consistent success of this approach and has remained a challenge for most LBPs under development. We recently demonstrated high-level engraftment of Bifidobacterium longum subsp. infantis (B. infantis) in adults when co-dosed with a specific prebiotic, human milk oligosaccharides (HMO). Here, we present a cellular kinetic-pharmacodynamic approach, analogous to pharmacokinetic-pharmacodynamic-based analyses of small molecule- and biologic-based drugs, to establish how HMO controls expansion, abundance, and metabolic output of B. infantis in a human microbiota-based model in gnotobiotic mice. Our data demonstrate that the HMO dose controls steady-state abundance of B. infantis in the microbiome, and that B. infantis together with HMO impacts gut metabolite levels in a targeted, HMO-dependent manner. We also found that HMO creates a privileged niche for B. infantis expansion across a 5-log range of bacterial inocula. These results demonstrate remarkable control of both B. infantis levels and the microbiome community metabolic outputs using this synbiotic approach, and pave the way for precision engineering of desirable microbes and metabolites to treat a range of diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Martin L. Lee
- Prolacta Bioscience, Duarte, CA, USA
- Department of Biostatistics, University of California Los Angeles Fielding School of Public Health, Los AngelesCA, USA
| | | | | | | |
Collapse
|
35
|
Yang S, Cai J, Su Q, Li Q, Meng X. Human milk oligosaccharides combine with Bifidobacterium longum to form the "golden shield" of the infant intestine: metabolic strategies, health effects, and mechanisms of action. Gut Microbes 2024; 16:2430418. [PMID: 39572856 PMCID: PMC11587862 DOI: 10.1080/19490976.2024.2430418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 08/04/2024] [Accepted: 11/12/2024] [Indexed: 11/26/2024] Open
Abstract
Human milk oligosaccharides (HMOs) are the third most important nutrient in human milk and are the gold standard for infant nutrition. Due to the lack of an enzyme system capable of utilizing HMOs in the infant intestine, HMOs cannot be directly utilized. Instead, they function as natural prebiotics, participating in the establishment of the intestinal microbiota as a "bifidus factor." A crucial colonizer of the early intestine is Bifidobacterium longum (B. longum), particularly its subspecies B. longum subsp. infantis, which is the most active consumer of HMOs. However, due to the structural diversity of HMOs and the specificity of B. longum strains, studies on their synergy are limited. An in-depth investigation into the mechanisms of HMO utilization by B. longum is essential for applying both as synbiotics to promote early intestinal development in infants. This review describes the colonization advantages of B. longum in the infant intestinal tract and its metabolic strategies for HMOs. It also summarizes recent studies on the effect and mechanism of B. longum and HMOs in infant intestinal development directly or indirectly through the action of metabolites. In conclusion, further structural analysis of HMOs and a deeper understanding of the interactions between B. longum and HMOs, as well as clinical trials, are necessary to lay the foundation for future practical applications as synbiotics.
Collapse
Affiliation(s)
- Shuo Yang
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Junwu Cai
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Qian Su
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Qiaohui Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Xiangchen Meng
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| |
Collapse
|
36
|
Li D, Lin Q, Luo F, Wang H. Insights into the Structure, Metabolism, Biological Functions and Molecular Mechanisms of Sialic Acid: A Review. Foods 2023; 13:145. [PMID: 38201173 PMCID: PMC10779236 DOI: 10.3390/foods13010145] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/18/2023] [Accepted: 12/26/2023] [Indexed: 01/12/2024] Open
Abstract
Sialic acid (SA) is a kind of functional monosaccharide which exists widely in edible bird's nest (EBN), milk, meat, mucous membrane surface, etc. SA is an important functional component in promoting brain development, anti-oxidation, anti-inflammation, anti-virus, anti-tumor and immune regulation. The intestinal mucosa covers the microbial community that has a significant impact on health. In the gut, SA can also regulate gut microbiota and metabolites, participating in different biological functions. The structure, source and physiological functions of SA were reviewed in this paper. The biological functions of SA through regulating key signaling pathways and target genes were discussed. In summary, SA can modulate gut microbiota and metabolites, which affect gene expressions and exert its biological activities. It is helpful to provide scientific reference for the further investigation of SA in the functional foods.
Collapse
Affiliation(s)
- Dan Li
- Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Processed Food for Special Medical Purpose, National Engineering Laboratory for Deep Process of Rice and Byproducts, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China;
- Hunan Engineering Research Center of Full Life-Cycle Energy-Efficient Buildings and Environmental Health, School of Civil Engineering, Central South University of Forestry and Technology, Changsha 410004, China
| | - Qinlu Lin
- Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Processed Food for Special Medical Purpose, National Engineering Laboratory for Deep Process of Rice and Byproducts, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China;
| | - Feijun Luo
- Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Processed Food for Special Medical Purpose, National Engineering Laboratory for Deep Process of Rice and Byproducts, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China;
| | - Hanqing Wang
- Hunan Engineering Research Center of Full Life-Cycle Energy-Efficient Buildings and Environmental Health, School of Civil Engineering, Central South University of Forestry and Technology, Changsha 410004, China
| |
Collapse
|
37
|
Wang A, Diana A, Rahmannia S, Gibson RS, Houghton LA, Slupsky CM. Impact of milk secretor status on the fecal metabolome and microbiota of breastfed infants. Gut Microbes 2023; 15:2257273. [PMID: 37741856 PMCID: PMC10519369 DOI: 10.1080/19490976.2023.2257273] [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: 03/20/2023] [Accepted: 09/06/2023] [Indexed: 09/25/2023] Open
Abstract
Maternal secretor status has been shown to be associated with the presence of specific fucosylated human milk oligosaccharides (HMOs), and the impact of maternal secretor status on infant gut microbiota measured through 16s sequencing has previously been reported. None of those studies have confirmed exclusive breastfeeding nor investigated the impact of maternal secretor status on gut microbial fermentation products. The present study focused on exclusively breastfed (EBF) Indonesian infants, with exclusive breastfeeding validated through the stable isotope deuterium oxide dose-to-mother (DTM) technique, and the impact of maternal secretor status on the infant fecal microbiome and metabolome. Maternal secretor status did not alter the within-community (alpha) diversity, between-community (beta) diversity, or the relative abundance of bacterial taxa at the genus level. However, infants fed milk from secretor (Se+) mothers exhibited a lower level of fecal succinate, amino acids and their derivatives, and a higher level of 1,2-propanediol when compared to infants fed milk from non-secretor (Se-) mothers. Interestingly, for infants consuming milk from Se+ mothers, there was a correlation between the relative abundance of Bifidobacterium and Streptococcus, and between each of these genera and fecal metabolites that was not observed in infants receiving milk from Se- mothers. Our findings indicate that the secretor status of the mother impacts the gut microbiome of the exclusively breastfed infant.
Collapse
Affiliation(s)
- Aidong Wang
- Department of Food Science and Technology, University of California, Davis, CA, USA
| | - Aly Diana
- Department of Public Health, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
- Nutrition Working Group, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Sofa Rahmannia
- Faculty of Medicine, Universitas Pasundan, Bandung, Indonesia
- School of Population and Global Health, University of Western Australia, Crawley, Western Australia, Australia
| | - Rosalind S Gibson
- Department of Human Nutrition, University of Otago, Dunedin, New Zealand
| | - Lisa A Houghton
- Department of Human Nutrition, University of Otago, Dunedin, New Zealand
| | - Carolyn M Slupsky
- Department of Food Science and Technology, University of California, Davis, CA, USA
- Department of Nutrition, University of California, Davis, CA, USA
| |
Collapse
|
38
|
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: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [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
|
39
|
Wang Y, Florez ID, Morgan RL, Foroutan F, Chang Y, Crandon HN, Zeraatkar D, Bala MM, Mao RQ, Tao B, Shahid S, Wang X, Beyene J, Offringa M, Sherman PM, El Gouhary E, Guyatt GH, Sadeghirad B. Probiotics, Prebiotics, Lactoferrin, and Combination Products for Prevention of Mortality and Morbidity in Preterm Infants: A Systematic Review and Network Meta-Analysis. JAMA Pediatr 2023; 177:1158-1167. [PMID: 37782505 PMCID: PMC10546299 DOI: 10.1001/jamapediatrics.2023.3849] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/01/2023] [Indexed: 10/03/2023]
Abstract
Importance Modulation of intestinal microbiome by administering probiotics, prebiotics, or both may prevent morbidity and mortality in premature infants. Objective To assess the comparative effectiveness of alternative prophylactic strategies through a network meta-analysis (NMA) of randomized clinical trials. Data Sources MEDLINE, EMBASE, Science Citation Index Expanded, CINAHL, Scopus, Cochrane CENTRAL, and Google Scholar from inception until May 10, 2023. Study Selection Eligible trials tested probiotics, prebiotics, lactoferrin, and combination products for prevention of morbidity or mortality in preterm infants. Data Extraction and Synthesis A frequentist random-effects model was used for the NMA, and the certainty of evidence and inferences regarding relative effectiveness were assessed using the GRADE approach. Main Outcomes and Measures All-cause mortality, severe necrotizing enterocolitis, culture-proven sepsis, feeding intolerance, time to reach full enteral feeding, and duration of hospitalization. Results A total of 106 trials involving 25 840 preterm infants were included. Only multiple-strain probiotics were associated with reduced all-cause mortality compared with placebo (risk ratio [RR], 0.69; 95% CI, 0.56 to 0.86; risk difference [RD], -1.7%; 95% CI, -2.4% to -0.8%). Multiple-strain probiotics alone (vs placebo: RR, 0.38; 95% CI, 0.30 to 0.50; RD, -3.7%; 95% CI, -4.1% to -2.9%) or in combination with oligosaccharides (vs placebo: RR, 0.13; 95% CI, 0.05 to 0.37; RD, -5.1%; 95% CI, -5.6% to -3.7%) were among the most effective interventions reducing severe necrotizing enterocolitis. Single-strain probiotics in combination with lactoferrin (vs placebo RR, 0.33; 95% CI, 0.14 to 0.78; RD, -10.7%; 95% CI, -13.7% to -3.5%) were the most effective intervention for reducing sepsis. Multiple-strain probiotics alone (RR, 0.61; 95% CI, 0.46 to 0.80; RD, -10.0%; 95% CI, -13.9% to -5.1%) or in combination with oligosaccharides (RR, 0.45; 95% CI, 0.29 to 0.67; RD, -14.1%; 95% CI, -18.3% to -8.5%) and single-strain probiotics (RR, 0.61; 95% CI, 0.51 to 0.72; RD, -10.0%; 95% CI, -12.6% to -7.2%) proved of best effectiveness in reduction of feeding intolerance vs placebo. Single-strain probiotics (MD, -1.94 days; 95% CI, -2.96 to -0.92) and multistrain probiotics (MD, -2.03 days; 95% CI, -3.04 to -1.02) proved the most effective in reducing the time to reach full enteral feeding compared with placebo. Only single-strain and multistrain probiotics were associated with greater effectiveness compared with placebo in reducing duration of hospitalization (MD, -3.31 days; 95% CI, -5.05 to -1.58; and MD, -2.20 days; 95% CI, -4.08 to -0.31, respectively). Conclusions and Relevance In this systematic review and NMA, moderate- to high-certainty evidence demonstrated an association between multistrain probiotics and reduction in all-cause mortality; these interventions were also associated with the best effectiveness for other key outcomes. Combination products, including single- and multiple-strain probiotics combined with prebiotics or lactoferrin, were associated with the largest reduction in morbidity and mortality.
Collapse
Affiliation(s)
- Yuting Wang
- Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University, Hamilton, Ontario, Canada
| | - Ivan D. Florez
- Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University, Hamilton, Ontario, Canada
- Department of Pediatrics, University of Antioquia, Medellin, Colombia
- Pediatric Intensive Care Unit, Clínica Las Americas-AUNA, Medellin, Colombia
| | - Rebecca L. Morgan
- Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University, Hamilton, Ontario, Canada
- School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Farid Foroutan
- Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University, Hamilton, Ontario, Canada
- Ted Rogers Centre for Heart Research, University Health Network, Toronto, Ontario, Canada
| | - Yaping Chang
- Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University, Hamilton, Ontario, Canada
| | - Holly N. Crandon
- Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, Ontario, Canada
| | - Dena Zeraatkar
- Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University, Hamilton, Ontario, Canada
- Department of Anesthesia, McMaster University, Hamilton, Ontario, Canada
| | - Malgorzata M. Bala
- Department of Hygiene and Dietetics, Jagiellonian University Medical College, Krakow, Poland
| | - Randi Q. Mao
- Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Brendan Tao
- Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Shaneela Shahid
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Xiaoqin Wang
- Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, Ontario, Canada
| | - Joseph Beyene
- Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University, Hamilton, Ontario, Canada
| | - Martin Offringa
- Child Health Evaluative Sciences, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Philip M. Sherman
- Division of Gastroenterology, Hepatology and Nutrition, Hospital for Sick Children, Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Enas El Gouhary
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Gordon H. Guyatt
- Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University, Hamilton, Ontario, Canada
| | - Behnam Sadeghirad
- Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, Ontario, Canada
- Department of Anesthesia, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
40
|
Dubernat L, Marousez L, Desseyn JL, Gouyer V, Hermann E, Gottrand F, Ley D, Lesage J. [Human milk oligosaccharides play major roles in child development and future health]. Med Sci (Paris) 2023; 39:869-875. [PMID: 38018931 DOI: 10.1051/medsci/2023164] [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: 11/30/2023] Open
Abstract
Human milk oligosaccharides (HMO) represent the third largest component of human breast milk (BM). The BM level is comprised between 5 to 20 g per liter and they have a great structural complexity with more than 150 HMO characterized to date. In this review, we present a summary of the main experimental and clinical data that have demonstrated their multiple biological roles in infants such as for gut development, microbiota, immune protection and neurodevelopment. Some HMO-enriched infant formulas are available yet, even if their benefits on the infant health remain to be confirmed. Further researches could allow therapeutic use in preterm newborns or in infants with intestinal diseases. Experimental data suggest that they could also be used in the prevention of some chronic diseases with immunometabolic or neurodevelopmental components.
Collapse
Affiliation(s)
- Laure Dubernat
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE, F-59000 Lille, France
| | - Lucie Marousez
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE, F-59000 Lille, France
| | - Jean-Luc Desseyn
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE, F-59000 Lille, France
| | - Valérie Gouyer
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE, F-59000 Lille, France
| | - Emmanuel Hermann
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE, F-59000 Lille, France
| | - Frédéric Gottrand
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE, F-59000 Lille, France - Service de gastroentérologie, hépatologie et nutrition, département de pédiatrie, hôpital Jeanne de Flandre, CHU Lille, F-59000 Lille, France
| | - Delphine Ley
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE, F-59000 Lille, France - Service de gastroentérologie, hépatologie et nutrition, département de pédiatrie, hôpital Jeanne de Flandre, CHU Lille, F-59000 Lille, France
| | - Jean Lesage
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE, F-59000 Lille, France
| |
Collapse
|
41
|
Nogacka AM, Cuesta I, Gueimonde M, de los Reyes-Gavilán CG. 2-Fucosyllactose Metabolism by Bifidobacteria Promotes Lactobacilli Growth in Co-Culture. Microorganisms 2023; 11:2659. [PMID: 38004671 PMCID: PMC10673426 DOI: 10.3390/microorganisms11112659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
Breastfeeding is recognized as the gold standard in infant nutrition, not only because of breastmilk's intrinsic nutritional benefits but also due to the high content of different bioactive components such as 2-fucosyllactose (2'FL) in the mother's milk. It promotes the growth of its two major consumers, Bifidobacterium longum ssp. infantis and Bifidobacterium bifidum, but the effect on other intestinal microorganisms of infant microbiota remains incompletely understood. pH-uncontrolled fecal cultures from infants donors identified as "fast 2'FL -degrader" microbiota phenotype were used for the isolation of 2'FL-associated microorganisms. The use of specific selective agents allowed the successful isolation of B. bifidum IPLA20048 and of Lactobacillus gasseri IPLA20136. The characterization of 2'FL consumption and its moieties has revealed more pronounced growth, pH drop, and lactic acid production after 2'FL consumption when both microorganisms were grown together. The results point to an association between B. bifidum IPLA20048 and L. gasseri IPLA20136 in which L. gasseri is able to use the galactose from the lactose moiety after the hydrolysis of 2'FL by B. bifidum. The additional screening of two groups of bifidobacteria (n = 38), fast and slow degraders of 2'FL, in co-culture with lactobacilli confirmed a potential cross-feeding mechanism based on degradation products released from bifidobacterial 2'FL break-down. Our work suggests that this phenomenon may be widespread among lactobacilli and bifidobacteria in the infant gut. More investigation is needed to decipher how the ability to degrade 2'FL and other human milk oligosaccharides could influence the microbiota establishment in neonates and the evolution of the microbiota in adult life.
Collapse
Affiliation(s)
- Alicja M. Nogacka
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (I.C.); (M.G.); (C.G.d.l.R.-G.)
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain
| | - Isabel Cuesta
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (I.C.); (M.G.); (C.G.d.l.R.-G.)
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (I.C.); (M.G.); (C.G.d.l.R.-G.)
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain
| | - Clara G. de los Reyes-Gavilán
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (I.C.); (M.G.); (C.G.d.l.R.-G.)
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain
| |
Collapse
|
42
|
Zhang L, Lin Q, Zhang J, Shi Y, Pan L, Hou Y, Peng X, Li W, Wang J, Zhou P. Qualitative and Quantitative Changes of Oligosaccharides in Human and Animal Milk over Lactation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:15553-15568. [PMID: 37815401 DOI: 10.1021/acs.jafc.3c03181] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/11/2023]
Abstract
The aim of this study was to investigate the changes in human and animal milk oligosaccharides over lactation. In total, 89, 97, 115, and 71 oligosaccharides were identified in human, bovine, goat, and camel milk. The number of common oligosaccharides between camel and human milk was the highest (16 and 17 in transitional and mature milk). With respect to the absolute concentration of eight oligosaccharides (2'-FL, 3-FL, α3'-GL, LNT, LNnT, 3'-SL, 6'-SL, and DSL), 2'-FL, 3'-FL, LNT, and LNnT were much higher in human than three animal species. 3'-SL had a similar concentration in bovine colostrum (322.2 μg/mL) and human colostrum (321.0 μg/mL), followed by goat colostrum (105.1 μg/mL); however, it had the highest concentration in camel mature milk (304.5 μg/mL). The ratio of 6'-SL and 3'-SL (1.77) in goat colostrum was similar to that in human colostrum (1.68), followed by bovine colostrum (0.13). In terms of changes of eight oligosaccharides over lactation, they all decreased with the increase of lactation in bovine and goat milk; however, α3'-GL, 2'-FL, and 3-FL increased in camel species, and LNT increased first and then decreased over lactation in human milk. This study provides a better understanding of the variation of milk oligosaccharides related to lactation and species.
Collapse
Affiliation(s)
- Lina Zhang
- State Key Laboratory of Food Science & Technology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
- School of Food Science & Technology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Qiaran Lin
- State Key Laboratory of Food Science & Technology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Jinyue Zhang
- State Key Laboratory of Food Science & Technology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Yue Shi
- State Key Laboratory of Food Science & Technology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Lina Pan
- Ausnutria Dairy (China) Company, Limited, Changsha, Hunan 410200, People's Republic of China
- National Center of Technology Innovation for Dairy, Hohhot, Inner Mongolia 010110, People's Republic of China
| | - Yanmei Hou
- Ausnutria Hyproca Nutrition Company, Limited, Changsha, Hunan 410011, People's Republic of China
| | - Xiaoyu Peng
- Ausnutria Dairy (China) Company, Limited, Changsha, Hunan 410200, People's Republic of China
| | - Wei Li
- Ausnutria Dairy (China) Company, Limited, Changsha, Hunan 410200, People's Republic of China
| | - Jiaqi Wang
- Ausnutria Dairy (China) Company, Limited, Changsha, Hunan 410200, People's Republic of China
| | - Peng Zhou
- State Key Laboratory of Food Science & Technology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
- School of Food Science & Technology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| |
Collapse
|
43
|
Jacobs JP, Lee ML, Rechtman DJ, Sun AK, Autran C, Niklas V. Human milk oligosaccharides modulate the intestinal microbiome of healthy adults. Sci Rep 2023; 13:14308. [PMID: 37652940 PMCID: PMC10471580 DOI: 10.1038/s41598-023-41040-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 08/18/2023] [Indexed: 09/02/2023] Open
Abstract
Human milk contains over 200 distinct oligosaccharides, which are critical to shaping the developing neonatal gut microbiome. To investigate whether a complex mixture of human milk oligosaccharides (HMOs) would similarly modulate the adult gut microbiome, HMO-Concentrate derived from pooled donor breast milk was administered orally to 32 healthy adults for 7 days followed by 21 days of monitoring. Fecal samples were collected for 16S rRNA gene sequencing, shotgun metagenomics, and metabolomics analyses. HMO-Concentrate induced dose-dependent Bifidobacterium expansion, reduced microbial diversity, and altered microbial gene content. Following HMO cessation, a microbial succession occurred with diverse taxonomic changes-including Bacteroides expansion-that persisted through day 28. This was associated with altered microbial gene content, shifts in serum metabolite levels, and increased circulating TGFβ and IL-10. Incubation of cultured adult microbiota with HMO-Concentrate induced dose-dependent compositional shifts that were not recapitulated by individual HMOs or defined mixtures of the 10 most abundant HMOs in HMO-Concentrate at their measured concentrations. These findings support that pooled donor HMOs can exert direct effects on adult gut microbiota and that complex mixtures including low abundance HMOs present in donor milk may be required for maximum effect.Registration: ClinicalTrials.gov NCT05516225.
Collapse
Affiliation(s)
- Jonathan P Jacobs
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
- Goodman-Luskin Microbiome Center, University of California Los Angeles, Los Angeles, CA, USA.
- Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, USA.
| | - Martin L Lee
- Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, USA
- Prolacta Bioscience, Duarte, CA, USA
| | | | | | | | - Victoria Niklas
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Oak Hill Bio Ltd, Altrincham, Cheshire WA14 2DT, United Kingdom
| |
Collapse
|
44
|
Zhao K, Pang H, Shao K, Yang Z, Li S, He N. The function of human milk oligosaccharides and their substitute oligosaccharides as probiotics in gut inflammation. Food Funct 2023; 14:7780-7798. [PMID: 37575049 DOI: 10.1039/d3fo02092d] [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: 08/15/2023]
Abstract
Gut inflammation seriously affects the healthy life of patients, and has a trend of increasing incidence rate. However, the current methods for treating gut inflammation are limited to surgery and drugs, which can cause irreversible damage to patients, especially infants. As natural oligosaccharides in human breast milk, human milk oligosaccharides (HMOs) function as probiotics in treating and preventing gut inflammation: improving the abundance of the gut microbiota, increasing the gut barrier function, and reducing the gut inflammatory reaction. Meanwhile, due to the complexity and high cost of their synthesis, people are searching for functional oligosaccharides that can replace HMOs as a food additive in infants milk powder and adjuvant therapy for chronic inflammation. The purpose of this review is to summarize the therapeutic and preventive effects of HMOs and their substitute functional oligosaccharides as probiotics in gut inflammation, and to summarize the prospect of their application in infant breast milk replacement in the future.
Collapse
Affiliation(s)
- Kunyi Zhao
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266003, China.
| | - Hao Pang
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266003, China.
| | - Kaidi Shao
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266003, China.
| | - Zizhen Yang
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266003, China.
| | - Shangyong Li
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266003, China.
| | - Ningning He
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266003, China.
| |
Collapse
|
45
|
Jackson PPJ, Wijeyesekera A, Rastall RA. Oligofructose alone and in combination with 2'fucosyllactose induces physiologically relevant changes in γ-aminobutyric acid and organic acid production compared to sole 2'fucosyllactose supplementation: an in vitro study. FEMS Microbiol Ecol 2023; 99:fiad100. [PMID: 37653466 PMCID: PMC10481994 DOI: 10.1093/femsec/fiad100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 08/10/2023] [Accepted: 08/25/2023] [Indexed: 09/02/2023] Open
Abstract
We explored the potential for the prebiotic oligofructose and prebiotic candidate 2'fucosyllactose, alone and in combination (50:50 blend) to induce physiologically relevant increases in neurotransmitter (γ-aminobutyric acid, serotonin, tryptophan, and dopamine) and organic acid (acetate, propionate, butyrate, lactate, and succinate) production as well as microbiome changes using anaerobic pH-controlled in vitro batch culture fermentations over 48 h. Changes in organic acid and neurotransmitter production were assessed by gas chromatography and liquid chromatography and, bacterial enumeration using fluorescence in situ hybridization, respectively. Both oligofructose and oligofructose/2'fucosyllactose combination fermentations induced physiologically relevant concentrations of γ-aminobutyric acid, acetate, propionate, butyrate, and succinate at completion (all P ≤ .05). A high degree of heterogeneity was seen amongst donors in both neurotransmitter and organic acid production in sole 2'FL fermentations suggesting a large responder/nonresponder status exists. Large increases in Bifidobacterium, Lactobacillus, and Bacteroides numbers were detected in oligofructose fermentation, smallest increases being detected in 2'fucosyllactose fermentation. Bacterial numbers in the combined oligofructose/2'fucosyllactose fermentation were closer to that of sole oligofructose. Our results indicate that oligofructose and oligofructose/2'fucosyllactose in combination have the potential to induce physiologically relevant increases in γ-aminobutyric and organic acid production along with offsetting the heterogenicity seen in response to sole 2'fucosyllactose supplementation.
Collapse
Affiliation(s)
- Peter Philip James Jackson
- Department of Food and Nutritional Sciences, University of Reading, Harry Nursten Building, Pepper Lane, Whiteknights, Reading RG6 6DZ, United Kingdom
| | - Anisha Wijeyesekera
- Department of Food and Nutritional Sciences, University of Reading, Harry Nursten Building, Pepper Lane, Whiteknights, Reading RG6 6DZ, United Kingdom
| | - Robert Adrian Rastall
- Department of Food and Nutritional Sciences, University of Reading, Harry Nursten Building, Pepper Lane, Whiteknights, Reading RG6 6DZ, United Kingdom
| |
Collapse
|
46
|
Jochum F, Meyer-Krott M, Hübler T, Lorenz M, Bedikian R, Zakarian J, Litzka A, Judex G, Hertzberg H, Klee D, Maurer L, Schacht M, Al-Radhi A, Maier J, Kröckel A, Faustmann C, Lavalle L, Dahbane S. Real-world evidence study on tolerance and growth in infants fed an infant formula with two human milk oligosaccharides vs mixed fed and exclusively breastfed infants. Mol Cell Pediatr 2023; 10:7. [PMID: 37597076 PMCID: PMC10439867 DOI: 10.1186/s40348-023-00162-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 08/04/2023] [Indexed: 08/21/2023] Open
Abstract
INTRODUCTION Human milk oligosaccharides (HMOs) are important components of human milk having diverse functions in the development of infants. Randomized controlled trials (RCTs) have demonstrated that infant formulas with the HMOs 2'-fucosyllactose (2'FL) and lacto-N-neotetraose (LNnT) are safe, well-tolerated, and support normal growth. This study aimed to generate real-world evidence (RWE) on growth and gastrointestinal (GI) tolerance in infants consuming a formula with 1 g/L 2'FL and 0.5 g/L LNnT, including a mixed feeding group not studied before in RCTs. PARTICIPANTS AND METHODS This 8-week open-label prospective multicenter study was conducted in Germany and Austria, and included groups of healthy, exclusively breastfed infants (BF), exclusively formula-fed infants (FF) who received the HMO-formula, and infants mixed fed with both HMO formula and human milk (MF). Co-primary outcomes were anthropometry and gastrointestinal tolerance via validated Infant Gastrointestinal Symptom Questionnaire (IGSQ). Secondary outcomes included formula satisfaction and adverse events (AEs). RESULTS One-hundred six infants completed the study (46 FF, 22 MF, and 38 BF). Mean anthropometric z-scores were comparable between groups and generally within ± 0.5 of WHO medians at week 8. IGSQ composite scores demonstrated good GI tolerance in all groups with no significant group differences at week 4 or 8. IGSQ composite scores in FF improved during the course of the study and parents provided high satisfaction ratings for the HMO-formula. Four potentially product-related AEs were reported in FF (no in MF). CONCLUSIONS In this RWE study examining an infant formula with HMOs, growth and GI tolerance outcomes were confirming the good tolerance and safety of this early feeding option previously reported in RCTs.
Collapse
Grants
- NCT05150288 Société des Produits Nestlé S.A., Vevey, Switzerland
- NCT05150288 Société des Produits Nestlé S.A., Vevey, Switzerland
- NCT05150288 Société des Produits Nestlé S.A., Vevey, Switzerland
- NCT05150288 Société des Produits Nestlé S.A., Vevey, Switzerland
- NCT05150288 Société des Produits Nestlé S.A., Vevey, Switzerland
- NCT05150288 Société des Produits Nestlé S.A., Vevey, Switzerland
- NCT05150288 Société des Produits Nestlé S.A., Vevey, Switzerland
- NCT05150288 Société des Produits Nestlé S.A., Vevey, Switzerland
- NCT05150288 Société des Produits Nestlé S.A., Vevey, Switzerland
- NCT05150288 Société des Produits Nestlé S.A., Vevey, Switzerland
- NCT05150288 Société des Produits Nestlé S.A., Vevey, Switzerland
- NCT05150288 Société des Produits Nestlé S.A., Vevey, Switzerland
- NCT05150288 Société des Produits Nestlé S.A., Vevey, Switzerland
- NCT05150288 Société des Produits Nestlé S.A., Vevey, Switzerland
- NCT05150288 Société des Produits Nestlé S.A., Vevey, Switzerland
- NCT05150288 Société des Produits Nestlé S.A., Vevey, Switzerland
- NCT05150288 Société des Produits Nestlé S.A., Vevey, Switzerland
- NCT05150288 Société des Produits Nestlé S.A., Vevey, Switzerland
Collapse
Affiliation(s)
- Frank Jochum
- Klinik für Kinder- und Jugendmedizin, Ev. Waldkrankenhaus Spandau Stadtrandstr. 555, 13589, Berlin und Medizinische Hochschule Brandenburg - Theodor Fontane (MHB), 16816, Neuruppin, Germany.
| | | | - Tina Hübler
- Gemeinschaftspraxis Kinder- und Jugendarztpraxis, Clemensstraße 4, 47608, Geldern, Germany
| | - Maja Lorenz
- Kinder- und Jugendarzt, Venloer Straße 67, 41751, Viersen, Germany
| | - Raffi Bedikian
- Kinder- und Jugendärztliche Gemeinschaftspraxis, Eugen-Zur-Nieden-Ring 1, 46145, Oberhausen, Germany
| | - Joseph Zakarian
- Kinderarztpraxis, Suitbertusstr. 31, 40223, Düsseldorf, Germany
| | - Anja Litzka
- Facharztpraxis für Kinder- und Jugendmedizin, Regensburger Str. 40, 93133, Burglengenfeld, Germany
| | - Guido Judex
- Zentrum für Kinder- und Jugendgesundheit Regensburg, Dr.-Leo-Ritter-Str. 4, 93049, Regensburg, Germany
| | - Holger Hertzberg
- Kinder- und Jugendarztpraxis, Ludwigstraße 4, 91126, Schwabach, Germany
| | - Daniela Klee
- Kinder- und Jugendarzt, Röntgen-Str. 6, 68642, Bürstadt, Germany
| | - Lothar Maurer
- Fachärzte für Säuglings-, Kinder- und Jugendmedizin, Welschgasse 39, 67227, Frankenthal, Germany
| | - Martin Schacht
- Facharzt für Säuglings-, Kinder- und Jugendmedizin, Schwachhauser Heerstr. 63a, 28211, Bremen, Germany
| | - Adnan Al-Radhi
- Kinder- und Jugendarzt Al-Radhi, Winckelhoferstrasse 3, 89584, Ehingen, Germany
| | - Jan Maier
- Kinder und Jugendarztpraxis, Geranienstr. 11, 70771, Leinfelden-Echterdingen, Germany
| | - Alexander Kröckel
- Kinder- und Jugendarztpraxis, Schwarzwurzelstraße 52/54, 12689, Berlin, Germany
| | - Christian Faustmann
- Facharzt für Kinder- und Jugendheilkunde, Wiener Strasse 8a, 7400, Oberwart, Austria
| | - Luca Lavalle
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Samir Dahbane
- Global Medical Affairs, Société des Produits Nestlé S.A., Vevey, Switzerland
| |
Collapse
|
47
|
Bozorgmehr T, Boutin RCT, Woodward SE, Donald K, Chow JM, Buck RH, Finlay BB. Early Life Exposure to Human Milk Oligosaccharides Reduces Allergic Response in a Murine Asthma Model. J Immunol Res 2023; 2023:9603576. [PMID: 37545544 PMCID: PMC10404156 DOI: 10.1155/2023/9603576] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/27/2023] [Accepted: 06/29/2023] [Indexed: 08/08/2023] Open
Abstract
Background Studies suggest that early-life gut microbiota composition and intestinal short-chain fatty acids (SCFAs) are linked to future asthma susceptibility. Furthermore, infancy offers a critical time window to modulate the microbiota and associated metabolites through diet-microbe interactions to promote infant health. Human milk oligosaccharides (HMOs), nondigestible carbohydrates abundant in breast milk, are prebiotics selectively metabolized by gut microbiota that consequently modify microbiome composition and SCFA production. Methods Using a house dust mite mouse model of allergy, we investigated the impacts of early oral treatment of pups with biologically relevant doses of 2'-fucosyllactose (2'-FL) and 6'-sialyllactose (6'-SL), two of the most abundant HMOs in human milk, in amelioration of allergic airway disease severity. Results We found that administration of 2'-FL and 6'-SL during early life reduced lung histopathology scores, circulating IgE, cytokine levels, and inflammatory cell infiltration, all hallmark symptoms of allergic asthma. HMO supplementation also increased the relative abundance of intestinal Bacteroidetes and Clostridia, known SCFA producers within the gut. Indeed, we detected increased SCFA concentrations in both the intestine and blood of adult mice who received HMOs prior to weaning. Conclusion We propose a model in which orally administered HMOs delivered during early life shift the microbiota toward increased production of SCFAs, which dampens the allergic immune responses behind allergy and asthma. Overall, these data suggest the potential for HMO supplementation to protect infants against asthma development later in life, with possible benefits against additional atopic diseases such as eczema and food allergies.
Collapse
Affiliation(s)
- Tahereh Bozorgmehr
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - Rozlyn C. T. Boutin
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Sarah E. Woodward
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Katherine Donald
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Jo May Chow
- Nutrition Division, Abbott Laboratories, Columbus, OH, USA
| | | | - B. Brett Finlay
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
48
|
Tarrant I, Finlay BB. Human milk oligosaccharides: potential therapeutic aids for allergic diseases. Trends Immunol 2023:S1471-4906(23)00111-4. [PMID: 37438187 DOI: 10.1016/j.it.2023.06.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/08/2023] [Accepted: 06/09/2023] [Indexed: 07/14/2023]
Abstract
Childhood allergy, including asthma, eczema, and food allergies, is a major global health burden, with prevalence increasing dramatically and novel interventions needed. Emerging research suggests that human milk oligosaccharides (HMOs), complex glycans found in breastmilk, have allergy-protective properties, indicating exciting therapeutic potential. This review evaluates current literature on the role of HMOs in allergy, assesses underlying immunological mechanisms, and discusses future research needed to translate findings into clinical implications. HMOs may mediate allergy risk through multiple structure-specific mechanisms, including microbiome modification, intestinal barrier maturation, immunomodulation, and gene regulation. Findings emphasize the importance of breastfeeding encouragement and HMO-supplemented formula milk for high allergy-risk infants. Although further investigation is necessary to determine the most efficacious structures against varying allergy phenotypes and their long-term efficacy, HMOs may represent a promising complementary tool for childhood allergy prevention.
Collapse
Affiliation(s)
- Isabel Tarrant
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - B Brett Finlay
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada; Department of Biochemistry, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
49
|
Durham SD, Wei Z, Lemay DG, Lange MC, Barile D. Creation of a milk oligosaccharide database, MilkOligoDB, reveals common structural motifs and extensive diversity across mammals. Sci Rep 2023; 13:10345. [PMID: 37365203 DOI: 10.1038/s41598-023-36866-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 06/13/2023] [Indexed: 06/28/2023] Open
Abstract
The carbohydrate fraction of most mammalian milks contains a variety of oligosaccharides that encompass a range of structures and monosaccharide compositions. Human milk oligosaccharides have received considerable attention due to their biological roles in neonatal gut microbiota, immunomodulation, and brain development. However, a major challenge in understanding the biology of milk oligosaccharides across other mammals is that reports span more than 5 decades of publications with varying data reporting methods. In the present study, publications on milk oligosaccharide profiles were identified and harmonized into a standardized format to create a comprehensive, machine-readable database of milk oligosaccharides across mammalian species. The resulting database, MilkOligoDB, includes 3193 entries for 783 unique oligosaccharide structures from the milk of 77 different species harvested from 113 publications. Cross-species and cross-publication comparisons of milk oligosaccharide profiles reveal common structural motifs within mammalian orders. Of the species studied, only chimpanzees, bonobos, and Asian elephants share the specific combination of fucosylation, sialylation, and core structures that are characteristic of human milk oligosaccharides. However, agriculturally important species do produce diverse oligosaccharides that may be valuable for human supplementation. Overall, MilkOligoDB facilitates cross-species and cross-publication comparisons of milk oligosaccharide profiles and the generation of new data-driven hypotheses for future research.
Collapse
Affiliation(s)
- Sierra D Durham
- Department of Food Science and Technology, University of California, Davis, One Shields Ave., Davis, CA, 95616, USA
| | - Zhe Wei
- Department of Food Science and Technology, University of California, Davis, One Shields Ave., Davis, CA, 95616, USA
| | - Danielle G Lemay
- Agricultural Research Service, U.S. Department of Agriculture, Western Human Nutrition Research Center, 430 West Health Sciences Dr., Davis, CA, 95616, USA
| | - Matthew C Lange
- International Center for Food Ontology Operability Data and Semantics, 216 F Street Ste. 139, Davis, CA, 95616, USA
| | - Daniela Barile
- Department of Food Science and Technology, University of California, Davis, One Shields Ave., Davis, CA, 95616, USA.
- Foods for Health Institute, University of California, Davis, One Shields Ave., Davis, CA, 95616, USA.
| |
Collapse
|
50
|
Yang L, Zhu Y, Meng J, Zhang W, Mu W. Recent progress in fucosylated derivatives of lacto- N-tetraose and lacto- N-neotetraose. Crit Rev Food Sci Nutr 2023; 64:10384-10396. [PMID: 37341681 DOI: 10.1080/10408398.2023.2224431] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2023]
Abstract
Human milk oligosaccharides (HMOs) have attracted considerable attention owing to their unique physiological functions. Two important tetrasaccharides, lacto-N-tetraose (LNT) and lacto-N-neotetraose (LNnT), are core structures of HMOs. Their safety has been evaluated and they can be added to infant formula as functional ingredients. The fucosylated derivatives of LNT and LNnT, mainly lacto-N-fucopentaose (LNFP) I, LNFP II, LNFP III, and lacto-N-difucohexaose I, exhibit prominent physiological characteristics, including modificating the intestinal microbiota, immunomodulation, anti-bacterial activities, and antiviral infection. However, they have received lesser attention than 2'-fucosyllactose. As precursors, LNT and LNnT are connected to one or two fucosyl units through α1,2/3/4 glycosidic bonds, forming a series of compounds with complex structures. These complex fucosylated oligosaccharides can be biologically synthesized using enzymatic and cell factory approaches. This review summarizes the occurrence, physiological effects, and biosynthesis of fucosylated LNT and LNnT derivatives and their future development.
Collapse
Affiliation(s)
- Longhao Yang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Yingying Zhu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Jiawei Meng
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Wenli Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Wanmeng Mu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|