1
|
Samarra A, Renwick S, Arzamasov AA, Rodionov DA, Spann K, Cabrera-Rubio R, Acuna-Gonzalez A, Martínez-Costa C, Hall L, Segata N, Osterman AL, Bode L, Collado MC. Human milk oligosaccharide metabolism and antibiotic resistance in early gut colonizers: insights from bifidobacteria and lactobacilli in the maternal-infant microbiome. Gut Microbes 2025; 17:2501192. [PMID: 40340669 PMCID: PMC12068340 DOI: 10.1080/19490976.2025.2501192] [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: 11/26/2024] [Revised: 03/22/2025] [Accepted: 04/28/2025] [Indexed: 05/10/2025] Open
Abstract
Breast milk, rich in human milk oligosaccharides (HMOs), supports the early-life colonization of beneficial bacteria such as bifidobacteria and lactobacilli, potentially reducing early-life antibiotic resistance. However, antibiotic treatment may interfere with the beneficial functions of HMO-degrading bacteria. This study investigated the metabolism of HMOs by bifidobacteria and lactobacilli isolated from human milk and mother-infant paired fecal samples, along with their antibiotic resistance profiles. Understanding these species- and sample-type-specific interactions will provide valuable insights into how bioactive components in human milk may shape the infant resistome during early life. A total of 39 Bifidobacterium and 14 Lactobacillaceae strains were isolated from paired mother-infant fecal and breast milk samples. Whole genome sequencing (WGS) allowed functional predictions on the HMO metabolism abilities and the resistance genotype of each strain. In vitro HMO utilization was assessed using growth kinetics assays combined with HMO glycoprofiling in culture supernatant. The minimum inhibitory concentration (MIC) was also determined for each strain. HMO metabolism by the bifidobacteria was species-specific. Bifidobacterium bifidum (B. bifidum) and Bifidobacterium longum subsp. infantis (B. infantis) exhibited the highest capacity for HMO degradation, consistent with genomic predictions. In contrast, lactobacilli were unable to degrade HMOs in vitro but were predicted to metabolize the by-products of HMO degradation. Phenotypic analysis revealed that B. bifidum strains had the lowest levels of antibiotic resistance, while Bifidobacterium animalis subsp. lactis (B. lactis) strains were resistant to most tested antibiotics. Overall, B. bifidum demonstrated the strongest HMO-degrading ability while remaining the most antibiotic-susceptible species. Early-life colonizing bifidobacterial species possess the essential machinery required to degrade HMOs and are highly susceptible to antibiotics. A better understanding of these dynamics could inform clinical strategies to protect and restore the infant gut microbiome, particularly in neonates exposed to antibiotics.
Collapse
Affiliation(s)
- Anna Samarra
- Department of Biotechnology, Institute of Agrochemistry and Food Technology- National Spanish Research Council (IATA-CSIC), Valencia, Spain
| | - Simone Renwick
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
- Mother-Milk-Infant Center of Research Excellence, University of California San Diego, La Jolla, CA, USA
| | - Aleksandr A. Arzamasov
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Dmitry A. Rodionov
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Kennedy Spann
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Raul Cabrera-Rubio
- Department of Biotechnology, Institute of Agrochemistry and Food Technology- National Spanish Research Council (IATA-CSIC), Valencia, Spain
| | - Antia Acuna-Gonzalez
- Food, Microbiome and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Cecilia Martínez-Costa
- Department of Pediatrics, School of Medicine, University of Valencia, Valencia, Spain
- Pediatric Gastroenterology and Nutrition Section, Hospital Clínico Universitario Valencia, Valencia, Spain
| | - Lindsay Hall
- Food, Microbiome and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- Department of Microbes, Infection and Microbiomes, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy
| | - Andrei L. Osterman
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Lars Bode
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
- Mother-Milk-Infant Center of Research Excellence, University of California San Diego, La Jolla, CA, USA
- Human Milk Institute, University of California San Diego, La Jolla, CA, USA
| | - MCarmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology- National Spanish Research Council (IATA-CSIC), Valencia, Spain
| |
Collapse
|
2
|
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 DOI: 10.3390/antibiotics14050488] [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: 03/27/2025] [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
- 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
| | - Caroline M Junker Mentzel
- Department of Veterinary and Animal Sciences, University of Copenhagen, Ridebanevej 9, DK-1870 Frederiksberg C, Denmark
| | - Sofie Kaas Lanng
- Department of Food Science, Aarhus University, Agro Food Park 48, DK-8200 Aarhus N, Denmark
| | - Elena Tina Gabriella Meloni
- Department of Food Science, University of Copenhagen, Rolighedsvej 26, DK-1958 Frederiksberg C, Denmark
- Department of Biotechnology and Biosciences BtBs, Piazza dell'Ateneo Nuovo, I-1-20126 Milan, Italy
| | | | - Camilla Hartmann Friis Hansen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Ridebanevej 9, DK-1870 Frederiksberg C, Denmark
| | - Axel Kornerup Hansen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Ridebanevej 9, DK-1870 Frederiksberg C, Denmark
| |
Collapse
|
3
|
Yu J, Li L, Kraithong S, Zou L, Zhang X, Huang R. Comprehensive review on human Milk oligosaccharides: Biosynthesis, structure, intestinal health benefits, immune regulation, neuromodulation mechanisms, and applications. Food Res Int 2025; 209:116328. [PMID: 40253162 DOI: 10.1016/j.foodres.2025.116328] [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: 12/06/2024] [Revised: 03/15/2025] [Accepted: 04/02/2025] [Indexed: 04/21/2025]
Abstract
This review provides a comprehensive analysis of the biosynthetic pathways of various oligosaccharides in Escherichia coli, structural characteristics, and bioactive mechanisms of human milk oligosaccharides (HMOs), with a particular emphasis on their roles in gut health, immune modulation, and neurodevelopment. HMOs primarily function as prebiotics, facilitating the growth of beneficial bacteria such as Bifidobacterium to maintain microbial homeostasis, with a discussion on the synergistic role of carbohydrate-binding modules (CBMs). In immune modulation, HMOs interact with lectins on immune and epithelial cells, influencing immune responses via pathways such as Toll-like receptors (TLRs). Additionally, HMOs have been linked to enhanced cognitive, motor, and language development in infants, influencing genes such as GABRB2, SLC1A7, GLRA4, and CHRM3. The review also examines commercially available HMO-containing products and highlights future research directions and potential applications in nutrition and healthcare.
Collapse
Affiliation(s)
- Jieting Yu
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Le Li
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Supaluck Kraithong
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China; Guangxi Key Laboratory of Marine Drugs, Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China.
| | - Lingshan Zou
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Xiaoyong Zhang
- University Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Riming Huang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
4
|
Sheng M, Liu Y, Zhu Y, Wang R, Zhang W, Mu W. Efficient Biosynthesis of Sialyllacto- N-tetraose a by a Metabolically Engineered Escherichia coli BL21(DE3) Strain. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:6820-6827. [PMID: 40036487 DOI: 10.1021/acs.jafc.4c12332] [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: 03/06/2025]
Abstract
Recently, the construction of metabolically engineered strains for the microbial synthesis of human milk oligosaccharides (HMOs) has attracted increasing attention. However, fewer efforts were made in the in vivo biosynthesis of complex HMOs, especially sialylated complex HMOs. In this study, we engineered Escherichia coli BL21(DE3) to efficiently produce sialyllacto-N-tetraose a (LST-a) efficiently. Three sequential glycosylation steps were introduced to construct the LST-a pathway, catalyzed by β1,3-N-acetylglucosaminylation, β1,3-galactosylation, and α2,3-sialylation. Pathway genes for cytidine 5'-monophospho (CMP)-N-acetylneuraminic acid (Neu5Ac) were introduced to support the sialylation donor supply. Production of LST-a was improved by deleting competitive genes of CMP-Neu5Ac synthesis, screening a more efficient α2,3-sialyltransferase, and combinatorial optimization of pathway gene expression. LST-a was finally produced with the titer of 1.235 and 4.85 g/L by shake-flask and fed-batch cultivation, respectively, demonstrating the feasibility of efficient microbial production of complex sialylated HMOs.
Collapse
Affiliation(s)
- Mian Sheng
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yuanlin Liu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yingying Zhu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Ruiyan Wang
- Bloomage Biotechnology Corp., Ltd., Jinan, Shandong 250010, People's Republic of China
| | - Wenli Zhang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wanmeng Mu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| |
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
|
Li C, Liu Z, Li M, Miao M, Zhang T. Review on bioproduction of sialylated human milk oligosaccharides: Synthesis methods, physiologic functions, and applications. Carbohydr Polym 2025; 352:123177. [PMID: 39843081 DOI: 10.1016/j.carbpol.2024.123177] [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: 11/02/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025]
Abstract
Human milk oligosaccharides (HMOs) are crucial for promoting neonatal health, with sialylated oligosaccharides, a significant subclass, offering a variety of health benefits such as prebiotic effects, anti-inflammatory and antimicrobial properties, antiviral defense, and cognitive development support. Among these, 3'-sialyllactose (3'-SL) and 6'-sialyllactose (6'-SL) have received "GRAS" status from the U.S. Food and Drug Administration and approval from the European Food Safety Authority for use as novel food additives in infant formula and supplements. This review focuses on the synthesis methods of sialylated human milk oligosaccharides (SHMOs), their functional properties, downstreaming developments and application technologies. Given the challenges associated with achieving sufficient availability for food and medical applications, the review emphasizes the viability and efficiency of various production strategies. The review also highlights recent research advancements and offers insights for optimizing large-scale production to support future applications in the food and pharmaceutical industries.
Collapse
Affiliation(s)
- Chenchen Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Zhu Liu
- Zhejiang Institute for Food and Drug Control, Hangzhou, 310052, Zhejiang, China
| | - Mengli Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Ming Miao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Tao Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
7
|
Li W, Yang J. Investigating the Anna Karenina principle of the breast microbiome. BMC Microbiol 2025; 25:81. [PMID: 39979818 PMCID: PMC11841003 DOI: 10.1186/s12866-024-03738-y] [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: 06/06/2024] [Accepted: 12/30/2024] [Indexed: 02/22/2025] Open
Abstract
The relationship between the microbiome and disease has long been a central focus of research in human microbiome. Inspired by Leo Tolstoy's dictum, the Anna Karenina Principle (AKP) offers a framework for understanding the complex dynamics of microbial communities in response to perturbations, suggesting that dysbiotic individuals exhibit greater variability/heterogeneity in their microbiome compared to healthy counterparts. While some studies have proved the alignment of microbiome responses to disease with the AKP effect, it remains uncertain whether the human breast microbiome responds similarly to breast disease. This study used beta-diversity and similarity in Hill numbers, along with shared species analysis (SSA), to explore this issue. We observed that during mastitis, changes in both the taxa richness and composition in the breast milk microbiome align with the AKP effect, while alterations in abundant taxa exhibit an anti-AKP effect. The response of breast tissue microbiome to breast cancer differs from that of milk microbiome to mastitis. Breast cancer induce anti-AKP effects in taxa richness, and non-AKP effects in common taxa and taxa composition. Overall, our findings identified different responses to breast diseases across taxa abundance in the breast microbiome. Mastitis primarily involves increasing the heterogeneity of rare taxa in the breast milk microbiome, while breast cancer associates with decreased dispersion of rare taxa in the tissue microbiome.
Collapse
Affiliation(s)
- Wendy Li
- College of Biological Sciences and Technology, Taiyuan Normal University, Jinzhong, 030600, China.
| | - Jinghui Yang
- Department of Pediatrics, The First People's Hospital of Yunnan Province, Kunming, 650032, China.
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China.
- Yunnan Province Clinical Research Center for Hematologic Disease, Kunming, 650032, China.
- Yunnan Province Clinical Center for Hematologic Disease, Kunming, 650032, China.
| |
Collapse
|
8
|
Cai R, Zhang J, Song Y, Liu X, Xu H. Research Progress on the Degradation of Human Milk Oligosaccharides (HMOs) by Bifidobacteria. Nutrients 2025; 17:519. [PMID: 39940377 PMCID: PMC11820314 DOI: 10.3390/nu17030519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 01/26/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025] Open
Abstract
The purpose of this study was to investigate the degradation mechanism of Bifidobacterium on breast milk oligosaccharides (HMOs) and its application in infant nutrition. The composition and characteristics of HMOs were introduced, and the degradation mechanism of HMOs by Bifidobacterium was described, including intracellular and extracellular digestion and species-specific differences. The interaction between Bifidobacterium and Bacteroides in the process of degrading HMOs and its effect on intestinal microecology were analyzed. The effects of HMO formula milk powder on the intestinal microbiota of infants were discussed, including simulating breast milk composition, regulating intestinal flora and immune function, infection prevention, and brain development. Finally, the research results are summarized, and future research directions are proposed to provide directions for research in the field of infant nutrition.
Collapse
Affiliation(s)
| | | | | | - Xiaoyong Liu
- School of Biological Science and Technology, University of Jinan, Jinan 250024, China; (R.C.); (J.Z.); (Y.S.)
| | - Huilian Xu
- School of Biological Science and Technology, University of Jinan, Jinan 250024, China; (R.C.); (J.Z.); (Y.S.)
| |
Collapse
|
9
|
Zhao Y, Liang S, Fu X, Guo Y, Wang Y, Wang J, Wang X, Wang Z, Tao H, Han B, Wang J. Anti-Inflammatory and Antidiarrheal Effects of Two Strains of Lactic Acid Bacteria Isolated from Healthy Pets on Escherichia coli K88-Induced Diarrhea in Mice. Microorganisms 2025; 13:239. [PMID: 40005605 PMCID: PMC11857690 DOI: 10.3390/microorganisms13020239] [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: 12/09/2024] [Revised: 01/17/2025] [Accepted: 01/19/2025] [Indexed: 02/27/2025] Open
Abstract
Lactic acid bacteria play a crucial role in maintaining the health of the host's gut microbiota. In this study, the anti-inflammatory properties of Limosilactobacillus reuteri LR20-6 and Lacticplantibacillus plantarum L272 were evaluated using a mouse model of diarrhea induced by Escherichia coli. We also investigated their effects on gut microbiota regulation. The results indicated that both Lacticplantibacillus plantarum and Limosilactobacillus reuteri could reduce inflammation by inhibiting the expression of inflammatory factors IL-6 and TNF-α and blocking the MyD88 and NF-kB/p65 signaling pathways. Additionally, after intervention with these strains, the relative abundance of Lactobacillus was significantly increased. This suggested that Lacticplantibacillus plantarum and Limosilactobacillus reuteri could mitigate the severity of E. coli-induced diarrhea and enhance the abundance of beneficial probiotics in the gut of animals.
Collapse
Affiliation(s)
- Ya Zhao
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (Y.Z.); (S.L.); (X.F.); (Y.G.); (Y.W.); (J.W.); (X.W.); (Z.W.); (H.T.)
| | - Shukun Liang
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (Y.Z.); (S.L.); (X.F.); (Y.G.); (Y.W.); (J.W.); (X.W.); (Z.W.); (H.T.)
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Xiaoxin Fu
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (Y.Z.); (S.L.); (X.F.); (Y.G.); (Y.W.); (J.W.); (X.W.); (Z.W.); (H.T.)
| | - Yaping Guo
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (Y.Z.); (S.L.); (X.F.); (Y.G.); (Y.W.); (J.W.); (X.W.); (Z.W.); (H.T.)
| | - Yu Wang
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (Y.Z.); (S.L.); (X.F.); (Y.G.); (Y.W.); (J.W.); (X.W.); (Z.W.); (H.T.)
| | - Jiaxue Wang
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (Y.Z.); (S.L.); (X.F.); (Y.G.); (Y.W.); (J.W.); (X.W.); (Z.W.); (H.T.)
| | - Xiumin Wang
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (Y.Z.); (S.L.); (X.F.); (Y.G.); (Y.W.); (J.W.); (X.W.); (Z.W.); (H.T.)
| | - Zhenlong Wang
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (Y.Z.); (S.L.); (X.F.); (Y.G.); (Y.W.); (J.W.); (X.W.); (Z.W.); (H.T.)
| | - Hui Tao
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (Y.Z.); (S.L.); (X.F.); (Y.G.); (Y.W.); (J.W.); (X.W.); (Z.W.); (H.T.)
| | - Bing Han
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (Y.Z.); (S.L.); (X.F.); (Y.G.); (Y.W.); (J.W.); (X.W.); (Z.W.); (H.T.)
| | - Jinquan Wang
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (Y.Z.); (S.L.); (X.F.); (Y.G.); (Y.W.); (J.W.); (X.W.); (Z.W.); (H.T.)
| |
Collapse
|
10
|
Sun W, Tao L, Qian C, Xue PP, Du SS, Tao YN. Human milk oligosaccharides: bridging the gap in intestinal microbiota between mothers and infants. Front Cell Infect Microbiol 2025; 14:1386421. [PMID: 39835278 PMCID: PMC11743518 DOI: 10.3389/fcimb.2024.1386421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 12/04/2024] [Indexed: 01/22/2025] Open
Abstract
Breast milk is an essential source of infant nutrition. It is also a vital determinant of the structure and function of the infant intestinal microbial community, and it connects the mother and infant intestinal microbiota. Human milk oligosaccharides (HMOs) are a critical component in breast milk. HMOs can reach the baby's colon entirely from milk and become a fermentable substrate for some intestinal microorganisms. HMOs can enhance intestinal mucosal barrier function and affect the intestinal function of the host through immune function, which has a therapeutic effect on specific infant intestinal diseases, such as necrotizing enterocolitis. In addition, changes in infant intestinal microbiota can reflect the maternal intestinal microbiota. HMOs are a link between the maternal intestinal microbiota and infant intestinal microbiota. HMOs affect the intestinal microbiota of infants and are related to the maternal milk microbiota. Through breastfeeding, maternal microbiota and HMOs jointly affect infant intestinal bacteria. Therefore, HMOs positively influence the establishment and balance of the infant microbial community, which is vital to ensure infant intestinal function. Therefore, HMOs can be used as a supplement and alternative therapy for infant intestinal diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Ying-na Tao
- Department of Traditional Chinese Medicine, Shanghai Fourth People’s Hospital
Affiliated to Tongji University, Shanghai, China
| |
Collapse
|
11
|
Łubiech K, Twarużek M, Sinkiewicz-Darol E, Martysiak-Żurowska D, Kusznierewicz B. Breast Milk as a Source of Prebiotic Human Milk Oligosaccharides and Bacteria from the Lactobacillaceae Family. Folia Biol (Praha) 2025; 71:44-53. [PMID: 40308202 DOI: 10.14712/fb2025071010044] [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: 05/02/2025]
Abstract
Breast milk, as the optimal food for infants and young children, contains all the components necessary for proper growth and development. It is a rich source of both essential nutrients and biologically active factors, making breast milk a unique food with scientifically proven health-promoting properties. Among the entire range of biologically active factors, breast milk microorganisms and prebiotic factors, in the form of breast milk oligosaccharides, occupy an important place. The aim of our research was to determine the occurrence of bacteria with probiotic potential, belonging to the Lactobacillaceae family, in the environment of breast milk and breast milk oligosaccharides. The study included 63 human milk samples from breastfeeding women at various stages of lactation. Microorganism identification based on culture tests and MALDI TOF/MS, macronutrient analysis using the MIRIS human milk analyser, as well as analysis of human milk oligosaccharides using ultra-high-performance liquid chromatography coupled with high-resolution mass spectrometry were performed. The results have shown that breast milk from different breastfeeding women is characterized by great diversity in terms of the presence of Lacto-bacillaceae bacteria in its microbiological composition. These bacteria were present in 22.2 % of the tested breast milk samples. Analysis of the human milk oligosaccharide profile revealed a slightly higher content of prebiotic factors in breast milk samples containing Lactobacillaceae, including 2'-fucosyllactose, oligosaccharide occurring in the highest amount in breast milk.
Collapse
Affiliation(s)
- Kataryzna Łubiech
- Department of Physiology and Toxicology, Faculty of Biological Sciences, Kazimierz Wielki University, Bydgoszcz, Poland.
| | - Magdalena Twarużek
- Department of Physiology and Toxicology, Faculty of Biological Sciences, Kazimierz Wielki University, Bydgoszcz, Poland.
| | - Elena Sinkiewicz-Darol
- Department of Physiology and Toxicology, Faculty of Biological Sciences, Kazimierz Wielki University, Bydgoszcz, Poland.
- Human Milk Bank, Ludwik Rydygier Provincial Polyclinical Hospital in Toruń, Toruń, Poland.
| | - Dorota Martysiak-Żurowska
- Department of Food Chemistry, Technology and Biotechnology, Chemical Faculty, Gdansk University of Technology, Gdańsk, Poland.
| | - Barbara Kusznierewicz
- Department of Food Chemistry, Technology and Biotechnology, Chemical Faculty, Gdansk University of Technology, Gdańsk, Poland.
| |
Collapse
|
12
|
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
|
13
|
Bao S, Shen T, Shabahang M, Bai G, Li L. Enzymatic Synthesis of Disialyllacto-N-Tetraose (DSLNT) and Related Human Milk Oligosaccharides Reveals Broad Siglec Recognition of the Atypical Neu5Acα2-6GlcNAc Motif. Angew Chem Int Ed Engl 2024; 63:e202411863. [PMID: 39223086 PMCID: PMC11631665 DOI: 10.1002/anie.202411863] [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: 06/24/2024] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 09/04/2024]
Abstract
Sialic acids (Sias) are ubiquitously expressed on all types of glycans, typically as terminating residues. They usually link to galactose, N-acetylgalactosamine, or other Sia residues, forming ligands of many glycan-binding proteins. An atypical linkage to the C6 of N-acetylglucosamine (GlcNAc) has been identified in human milk oligosaccharides (HMOs, e.g., DSLNT) and tumor-associated glycoconjugates. Herein, describe the systematic synthesis of these HMOs in an enzymatic modular manner. The synthetic strategy relies on a novel activity of ST6GalNAc6 for efficient construction of the Neu5Acα2-6GlcNAc linkage, and another 12 specific enzyme modules for sequential HMO assembly. The structures enabled comprehensive exploration of their structure-function relationships using glycan microarrays, revealing broad yet distinct recognition by Siglecs of the atypical Neu5Acα2-6GlcNAc motif. The work provides tools and new insight for the functional study and potential applications of Siglecs and HMOs.
Collapse
Affiliation(s)
- Shumin Bao
- Department of Chemistry and Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - Tangliang Shen
- Department of Chemistry and Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - MohammadHossein Shabahang
- Department of Chemistry and Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - Guitao Bai
- Department of Chemistry and Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - Lei Li
- Department of Chemistry and Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
14
|
Natal ACDC, de Paula Menezes R, de Brito Röder DVD. Role of maternal milk in providing a healthy intestinal microbiome for the preterm neonate. Pediatr Res 2024:10.1038/s41390-024-03751-x. [PMID: 39663425 DOI: 10.1038/s41390-024-03751-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 10/28/2024] [Accepted: 11/10/2024] [Indexed: 12/13/2024]
Abstract
The immature gastrointestinal tract of preterm neonates leads to a delayed and distinctive establishment of the gut microbiome, making them susceptible to potentially pathogenic bacteria and increasing the risk of infections. Maternal milk, recognized as the optimal source of nutrition, plays a multifaceted role in modulating the gut microbiome of premature newborns. Human milk oligosaccharides, acting as prebiotics, provide essential nourishment for key bacteria such as Bifidobacterium, contributing to the proliferation of beneficial bacterial populations. Additionally, maternal milk is rich in Immunoglobulins that stimulate immune cell responses, providing protective effects on the infant's gut mucosa. Moreover, bioactive proteins such as secretory immunoglobulin A (SIgA), lactoferrin, lysozyme, and mucins play a crucial role in defending against pathogens and regulating the immune system at the cellular level. These proteins contribute not only to infection prevention but also emphasize the impact of breast milk in fortifying the body's innate defenses. This multifaceted role of maternal milk, including essential nutrients, beneficial bacteria, and bioactive proteins, highlights the importance of promoting the mother's own milk feeding in the Neonatal Intensive Care Unit (NICU). It not only optimizes the long-term outcomes and well-being of preterm infants but also provides a holistic approach to their health and development. IMPACT: This article contributes to the current understanding of the relationship between breastfeeding and the intestinal microbiota. Fill gaps in existing literature about the subject. Provides new insights for future research.
Collapse
Affiliation(s)
- Ana Catarina de Castro Natal
- Undergraduate Nursing, Faculty of Medicine (FAMED), Federal University of Uberlandia UFU, Uberlandia, MG, Brazil.
| | | | | |
Collapse
|
15
|
Jiang S, Qin J, Shi L, Feng J, Mo J, Su W, Cheng Y, Lv J, Li Q, Li S, Zeng L, Han B, Zhou J. Association among Gestational Weight Gain, Fucosylated Human Milk Oligosaccharides, and Breast Milk Microbiota─An Evidence in Healthy Mothers from Northwest China. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:25135-25145. [PMID: 39476856 PMCID: PMC11565758 DOI: 10.1021/acs.jafc.4c07050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 11/14/2024]
Abstract
This study investigates the relationship among maternal secretor status, human milk oligosaccharides (HMOs), and the composition of breastmilk microbiota in a cohort of healthy mothers from Shaanxi province, China. The results demonstrated that 78.9% of the mothers were secretors, exhibiting an active fucosyltransferase 2 gene (fut2) and producing α-1,2 fucosylated HMOs, which significantly affected the HMO profile. Secretor mothers had higher levels of 2'-FL and LNFPI in contrast to nonsecretors who displayed high levels of 3'-FL, LNFPII, and LNT. Furthermore, secretor mothers exhibited greater diversity in HMOs compared with nonsecretors, although no significant differences were observed in the breast milk microbiota composition. A correlation was identified between specific HMOs (2'-FL, 3'-FL, 6'-SL, and LNFPI) and the microbiota composition. Notably, mothers with normal weight gain during pregnancy demonstrated higher microbial diversity, with increased abundance of beneficial genera such as Bifidobacterium, Lactobacillus, and Ligilactobacillus. These findings contribute to the development of potential guidelines for providing personalized nutrition.
Collapse
Affiliation(s)
- Sijin Jiang
- School
of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Jiale Qin
- School
of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Lu Shi
- School
of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Jiayu Feng
- School
of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Jianhui Mo
- School
of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Wanghong Su
- School
of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Yue Cheng
- School
of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Jia Lv
- School
of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
- Department
of Pediatrics, The Second Affiliated Hospital
of Xi’an Jiaotong University, Xi’an, Shaanxi 710004, China
| | - Qiang Li
- School
of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Shaoru Li
- School
of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Lingxia Zeng
- School
of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Bei Han
- School
of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Jing Zhou
- Department
of Pediatrics, The Second Affiliated Hospital
of Xi’an Jiaotong University, Xi’an, Shaanxi 710004, China
| |
Collapse
|
16
|
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
|
17
|
Horwell E, Bearn P, Cutting SM. A microbial symphony: a literature review of the factors that orchestrate the colonization dynamics of the human colonic microbiome during infancy and implications for future health. MICROBIOME RESEARCH REPORTS 2024; 4:1. [PMID: 40207275 PMCID: PMC11977369 DOI: 10.20517/mrr.2024.32] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
Since the advent of new sequencing and bioinformatic technologies, our understanding of the human microbiome has expanded rapidly over recent years. Numerous studies have indicated causal links between alterations to the microbiome and a range of pathological conditions. Furthermore, a large body of epidemiological data is starting to suggest that exposure, or lack thereof, to specific microbial species during the first five years of life has key implications for long-term health outcomes. These include chronic inflammatory and metabolic conditions such as diabetes, asthma, inflammatory bowel disease (IBD), and obesity, with the effects lasting into adulthood. Human microbial colonisation during these first five years of life is a highly dynamic process, with multiple environmental exposures recently being characterised to have influence before the microbiome stabilises and resembles that of an adult at 3-5 years. This short period of time, known as the window of opportunity, appears to "prime" immunoregulation for later life. Understanding and appreciating this aspect of human physiology is therefore crucial for clinicians, scientists, and public health officials. This review outlines the most recent evidence for the pre- and post-natal environments that order the development of the microbiome, how these influences metabolic and immunoregulatory pathways, and their associated health outcomes. It also discusses the limitations of the current knowledge base, and describes the potential microbiome-mediated interventions and public health measures that may have therapeutic potential in the future.
Collapse
Affiliation(s)
- Edward Horwell
- Department of Biomedical Sciences, The Bourne Laboratory, Royal Holloway University of London, London TW20 0EX, UK
- Department of Colorectal Surgery, Ashford and Saint Peter’s NHS Foundation Trust, London KT16 0PZ, UK
| | - Philip Bearn
- Department of Colorectal Surgery, Ashford and Saint Peter’s NHS Foundation Trust, London KT16 0PZ, UK
| | - Simon M. Cutting
- Department of Biomedical Sciences, The Bourne Laboratory, Royal Holloway University of London, London TW20 0EX, UK
| |
Collapse
|
18
|
Borewicz K, Hornung B, Gu F, van der Zaal PH, Schols HA, Schaap PJ, Smidt H. Metatranscriptomic analysis indicates prebiotic effect of isomalto/malto-polysaccharides on human colonic microbiota in-vitro. Sci Rep 2024; 14:18866. [PMID: 39143192 PMCID: PMC11324910 DOI: 10.1038/s41598-024-69685-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 08/07/2024] [Indexed: 08/16/2024] Open
Abstract
Isomalto/malto-polysaccharides (IMMPs) are a novel type of soluble dietary fibres with a prebiotic potential promoting growth of beneficial microbes in the gut. However, the mode of action of IMMPs remains unknown. Previous studies on IMMPs showed an increase in total bacteria, especially lactobacilli, and higher production of short chain fatty acids (SCFA) when IMMPs were fed to rats or used during in vitro fermentation. Here we used metatranscriptomics to investigate how IMMPs with different amounts of α - (1 → 6) glycosidic linkages affected microbial function during incubation with human fecal inoculum. We showed that active microbial community dynamics during fermentation varied depending on the type of IMMP used and that the observed changes were reflected in the community gene expression profiles. Based on metatranscriptome analysis, members of Bacteroides, Lactobacillus and Bifidobacterium were the predominant degraders of IMMPs, and the increased gene expression in these bacteria correlated with high amounts of α - (1 → 6) glycosidic linkages. We also noted an increase in relative abundance of these bacteria and an activation of pathways involved in SCFA synthesis. Our findings could provide a baseline for more targeted approaches in designing prebiotics for specific bacteria and to achieve more controlled modulation of microbial activity towards desired health outcomes.
Collapse
Affiliation(s)
- Klaudyna Borewicz
- Laboratory of Microbiology, Wageningen University & Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
- Mead Johnson, Middenkampweg 2, 6545 CJ, Nijmegen, The Netherlands
| | - Bastian Hornung
- Laboratory of Microbiology, Wageningen University & Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands.
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands.
- CBG-MEB, Graadt Van Roggenweg 500, 3531AH, Utrecht, The Netherlands.
| | - Fangjie Gu
- Laboratory of Food Chemistry, Wageningen University & Research, Bornse Weilanden 9, 6708 WG, Wageningen, The Netherlands
- TUMCREATE, 1 CREATE Way, CREATE Tower, #10-02, Singapore, 138602, Singapore
| | - Pieter H van der Zaal
- Biobased Chemistry and Technology, Wageningen University & Research, Bornse Weilanden 9, 6708 WG, Wageningen, The Netherlands
- IFF, Willem Einthovenstraat 4, 2342 BH, Oegstgeest, The Netherlands
| | - Henk A Schols
- Laboratory of Food Chemistry, Wageningen University & Research, Bornse Weilanden 9, 6708 WG, Wageningen, The Netherlands
| | - Peter J Schaap
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Hauke Smidt
- Laboratory of Microbiology, Wageningen University & Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| |
Collapse
|
19
|
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
|
20
|
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
|
21
|
Ermann Lundberg L, Pallabi Mishra P, Liu P, Forsberg MM, Sverremark-Ekström E, Grompone G, Håkansson S, Linninge C, Roos S. Bifidobacterium longum subsp. longum BG-L47 boosts growth and activity of Limosilactobacillus reuteri DSM 17938 and its extracellular membrane vesicles. Appl Environ Microbiol 2024; 90:e0024724. [PMID: 38888338 PMCID: PMC11267924 DOI: 10.1128/aem.00247-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/14/2024] [Indexed: 06/20/2024] Open
Abstract
The aim of this study was to identify a Bifidobacterium strain that improves the performance of Limosilactobacillus reuteri DSM 17938. Initial tests showed that Bifidobacterium longum subsp. longum strains boosted the growth of DSM 17938 during in vivo-like conditions. Further characterization revealed that one of the strains, BG-L47, had better bile and acid tolerance compared to BG-L48, as well as mucus adhesion compared to both BG-L48 and the control strain BB536. BG-L47 also had the capacity to metabolize a broad range of carbohydrates and sugar alcohols. Mapping of glycoside hydrolase (GH) genes of BG-L47 and BB536 revealed many GHs associated with plant-fiber utilization. However, BG-L47 had a broader phenotypic fiber utilization capacity. In addition, B. longum subsp. longum cells boosted the bioactivity of extracellular membrane vesicles (MV) produced by L. reuteri DSM 17938 during co-cultivation. Secreted 5'-nucleotidase (5'NT), an enzyme that converts AMP into the signal molecule adenosine, was increased in MV boosted by BG-L47. The MV exerted an improved antagonistic effect on the pain receptor transient receptor potential vanilloid 1 (TRPV1) and increased the expression of the immune development markers IL-6 and IL-1ß in a peripheral blood mononuclear cell (PBMC) model. Finally, the safety of BG-L47 was evaluated both by genome safety assessment and in a human safety study. Microbiota analysis showed that the treatment did not induce significant changes in the composition. In conclusion, B. longum subsp. longum BG-L47 has favorable physiological properties, can boost the in vitro activity of L. reuteri DSM 17938, and is safe for consumption, making it a candidate for further evaluation in probiotic studies. IMPORTANCE By using probiotics that contain a combination of strains with synergistic properties, the likelihood of achieving beneficial interactions with the host can increase. In this study, we first performed a broad screening of Bifidobacterium longum subsp. longum strains in terms of synergistic potential and physiological properties. We identified a superior strain, BG-L47, with favorable characteristics and potential to boost the activity of the known probiotic strain Limosilactobacillus reuteri DSM 17938. Furthermore, we demonstrated that BG-L47 is safe for consumption in a human randomized clinical study and by performing a genome safety assessment. This work illustrates that bacteria-bacteria interactions differ at the strain level and further provides a strategy for finding and selecting companion strains of probiotics.
Collapse
Affiliation(s)
- Ludwig Ermann Lundberg
- Department of Molecular Sciences, Uppsala BioCenter, Swedish University of Agricultural Sciences, Uppsala, Sweden
- BioGaia, Stockholm, Sweden
| | - Punya Pallabi Mishra
- Department of Molecular Sciences, Uppsala BioCenter, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | | | - Manuel Mata Forsberg
- The Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Eva Sverremark-Ekström
- The Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | | | - Sebastian Håkansson
- BioGaia, Stockholm, Sweden
- Division of Applied Microbiology, Department of Chemistry, Lund University, Lund, Sweden
| | - Caroline Linninge
- BioGaia, Stockholm, Sweden
- Department of Food Technology, Engineering and Nutrition, Lund University, Lund, Sweden
| | - Stefan Roos
- Department of Molecular Sciences, Uppsala BioCenter, Swedish University of Agricultural Sciences, Uppsala, Sweden
- BioGaia, Stockholm, Sweden
| |
Collapse
|
22
|
Martínez Gascueña A, Wu H, Wang R, Owen CD, Hernando PJ, Monaco S, Penner M, Xing K, Le Gall G, Gardner R, Ndeh D, Urbanowicz PA, Spencer DIR, Walsh M, Angulo J, Juge N. Exploring the sequence-function space of microbial fucosidases. Commun Chem 2024; 7:137. [PMID: 38890439 PMCID: PMC11189522 DOI: 10.1038/s42004-024-01212-4] [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: 02/15/2024] [Accepted: 05/28/2024] [Indexed: 06/20/2024] Open
Abstract
Microbial α-L-fucosidases catalyse the hydrolysis of terminal α-L-fucosidic linkages and can perform transglycosylation reactions. Based on sequence identity, α-L-fucosidases are classified in glycoside hydrolases (GHs) families of the carbohydrate-active enzyme database. Here we explored the sequence-function space of GH29 fucosidases. Based on sequence similarity network (SSN) analyses, 15 GH29 α-L-fucosidases were selected for functional characterisation. HPAEC-PAD and LC-FD-MS/MS analyses revealed substrate and linkage specificities for α1,2, α1,3, α1,4 and α1,6 linked fucosylated oligosaccharides and glycoconjugates, consistent with their SSN clustering. The structural basis for the substrate specificity of GH29 fucosidase from Bifidobacterium asteroides towards α1,6 linkages and FA2G2 N-glycan was determined by X-ray crystallography and STD NMR. The capacity of GH29 fucosidases to carry out transfucosylation reactions with GlcNAc and 3FN as acceptors was evaluated by TLC combined with ESI-MS and NMR. These experimental data supported the use of SSN to further explore the GH29 sequence-function space through machine-learning models. Our lightweight protein language models could accurately allocate test sequences in their respective SSN clusters and assign 34,258 non-redundant GH29 sequences into SSN clusters. It is expected that the combination of these computational approaches will be used in the future for the identification of novel GHs with desired specificities.
Collapse
Affiliation(s)
- Ana Martínez Gascueña
- The Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
| | - Haiyang Wu
- The Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
- GuangDong Engineering Technology Research Center of Enzyme and Biocatalysis, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
| | - Rui Wang
- Beijing Key Lab of Traffic Data Analysis and Mining, Beijing Jiaotong University, Beijing, China
- Collaborative Innovation Center of Railway Traffic Safety, Beijing Jiaotong University, Beijing, China
- School of Computer and Information Technology, Beijing Jiaotong University, Beijing, China
| | - C David Owen
- Diamond Light Source Ltd, Diamond House, Harwell Science and Innovation Campus, Didcot, OX11 0FA, UK
- Research Complex at Harwell, Rutherford Appleton Laboratory, Harwell Oxford, Didcot, OX11 0FA, UK
| | - Pedro J Hernando
- The Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
- Iceni Glycoscience Ltd., Norwich Research Park, Norwich, NR4 7JG, UK
| | - Serena Monaco
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Matthew Penner
- Diamond Light Source Ltd, Diamond House, Harwell Science and Innovation Campus, Didcot, OX11 0FA, UK
- Research Complex at Harwell, Rutherford Appleton Laboratory, Harwell Oxford, Didcot, OX11 0FA, UK
| | - Ke Xing
- School of Computer and Information Technology, Beijing Jiaotong University, Beijing, China
| | - Gwenaelle Le Gall
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | | | - Didier Ndeh
- The Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
- University of Dundee, School of Life Sciences, Dundee, DD1 5EH, Scotland, UK
| | | | | | - Martin Walsh
- Diamond Light Source Ltd, Diamond House, Harwell Science and Innovation Campus, Didcot, OX11 0FA, UK
- Research Complex at Harwell, Rutherford Appleton Laboratory, Harwell Oxford, Didcot, OX11 0FA, UK
| | - Jesus Angulo
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
- Departamento de Química Orgánica, Universidad de Sevilla, 41012, Sevilla, Spain
- Instituto de Investigaciones Químicas (CSIC-US), 41092, Sevilla, Spain
| | - Nathalie Juge
- The Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK.
| |
Collapse
|
23
|
Matera M. Bifidobacteria, Lactobacilli... when, how and why to use them. GLOBAL PEDIATRICS 2024; 8:100139. [DOI: 10.1016/j.gpeds.2024.100139] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
24
|
Wong CB, Huang H, Ning Y, Xiao J. Probiotics in the New Era of Human Milk Oligosaccharides (HMOs): HMO Utilization and Beneficial Effects of Bifidobacterium longum subsp. infantis M-63 on Infant Health. Microorganisms 2024; 12:1014. [PMID: 38792843 PMCID: PMC11124435 DOI: 10.3390/microorganisms12051014] [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: 04/24/2024] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
A healthy gut microbiome is crucial for the immune system and overall development of infants. Bifidobacterium has been known to be a predominant species in the infant gut; however, an emerging concern is the apparent loss of this genus, in particular, Bifidobacterium longum subsp. infantis (B. infantis) in the gut microbiome of infants in industrialized nations, underscoring the importance of restoring this beneficial bacterium. With the growing understanding of the gut microbiome, probiotics, especially infant-type human-residential bifidobacteria (HRB) strains like B. infantis, are gaining prominence for their unique ability to utilize HMOs and positively influence infant health. This article delves into the physiology of a probiotic strain, B. infantis M-63, its symbiotic relationship with HMOs, and its potential in improving gastrointestinal and allergic conditions in infants and children. Moreover, this article critically assesses the role of HMOs and the emerging trend of supplementing infant formulas with the prebiotic HMOs, which serve as fuel for beneficial gut bacteria, thereby emulating the protective effects of breastfeeding. The review highlights the potential of combining B. infantis M-63 with HMOs as a feasible strategy to improve health outcomes in infants and children, acknowledging the complexities and requirements for further research in this area.
Collapse
Affiliation(s)
- Chyn Boon Wong
- International Division, Morinaga Milk Industry Co., Ltd., 5-2, Higashi Shimbashi 1-Chome, Minato-ku, Tokyo 105-7122, Japan
| | - Huidong Huang
- Nutrition Research Institute, Junlebao Dairy Group Co., Ltd., 36 Shitong Road, Shijiazhuang 050221, China
| | - Yibing Ning
- Nutrition Research Institute, Junlebao Dairy Group Co., Ltd., 36 Shitong Road, Shijiazhuang 050221, China
| | - Jinzhong Xiao
- Morinaga Milk Industry (Shanghai) Co., Ltd., Room 509 Longemont Yes Tower, No. 369 Kaixuan Road, Changning District, Shanghai 200050, China
- Department of Microbiota Research, Graduate School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Research Center for Probiotics, Department of Nutrition and Health, China Agricultural University, Beijing 100093, China
| |
Collapse
|
25
|
Tao M, Yang L, Zhao C, Zhao M, Zhang W, Zhu Y, Mu W. Implementation of a Quorum-Sensing System for Highly Efficient Biosynthesis of Lacto- N-neotetraose in Engineered Escherichia coli MG1655. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:7179-7186. [PMID: 38520358 DOI: 10.1021/acs.jafc.3c09162] [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: 03/25/2024]
Abstract
Lacto-N-neotetraose (LNnT), a prominent neutral human milk oligosaccharide (HMO), serves as a pivotal structural element in complex HMO biosynthesis. Given its promising health effects for infants, the biosynthesis of LNnT is garnering greater interest. Using a previously engineered strain as a chassis, a highly effective LNnT producer was constructed. First, LNnT synthesis in Escherichia coli MG1655 was achieved by introducing β1,3-N-acetylglucosaminyltransferase LgtA and β1,4-galactosyltransferase CpsIaJ, coupled with the optimization of enzyme expression levels using various promoters. Subsequently, ugd underwent disruption, and the galE gene was enhanced by replacing its promoter with PJ23119 or Ptac. Then, a lux-type quorum sensing (QS) system was applied to achieve varied metabolic regulation. Additionally, systematic optimization of the QS promoters was conducted to further improve the LNnT titer in the shake flask. Finally, the extracellular titer of LNnT was 20.33 g/L, accompanied by a productivity of 0.41 g/L/h.
Collapse
Affiliation(s)
- Mengting Tao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Longhao Yang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Chunhua Zhao
- Bloomature Biotechnology Corporation, Limited, Beijing 102629, People's Republic of China
| | - Mingli Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Wenli Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Yingying Zhu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Wanmeng Mu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| |
Collapse
|
26
|
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
|
27
|
Chen C, Xu J, Han T, Chen G, Yu K, Du C, Shen W, Sun Y, Zeng X. Microencapsulation as a Protective Strategy for Sialylated Immunoglobulin G: Efficacy in Alleviating Symptoms of Dextran Sulfate Sodium-Induced Colitis in Mice and Potential Mechanisms. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:4074-4088. [PMID: 38323407 DOI: 10.1021/acs.jafc.3c07733] [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: 02/08/2024]
Abstract
Sialylated immunoglobulin G (IgG) is a vital glycoprotein in breast milk with the ability to promote the growth of Bifidobacterium in gut microbiota and relieve inflammatory bowel disease (IBD) symptoms in vitro. Here, it was found that the microcapsules with sialylated IgG could protect and release sialylated IgG with its structure and function in the intestine. Furthermore, the sialylated IgG microcapsules alleviated the clinical symptoms (body weight, feed quantity, and colon length loss), decreased disease activity index score, suppressed the production of pro-inflammatory cytokines (TNF-α, IL-6, IL-1β, IFN-γ, and MCP-1) and endotoxin (lipopolysaccharide), and enhanced the intestinal mucosal barrier (Claudin1, Muc2, Occludin, and ZO-1) in dextran sulfate sodium (DSS)-induced colitis mice. Additionally, the sialylated IgG microcapsules improved the gut microbiota by increasing the relative abundance of critical microbe Bifidobacterium bifidum and promoted the production of short-chain fatty acids (SCFAs). Correlation analysis indicated that the key microbes were strongly correlated with pro-inflammatory factors, clinical symptoms, tight junction protein, and SCFAs. These findings suggest that the sialylated IgG microcapsules have the potential to be used as a novel therapeutic approach for treating IBD.
Collapse
Affiliation(s)
- Chunxu Chen
- College of Food Engineering, Anhui Science and Technology University, Fengyang 233100, Anhui, China
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Jiaming Xu
- College of Food Engineering, Anhui Science and Technology University, Fengyang 233100, Anhui, China
| | - Tianxiang Han
- College of Food Engineering, Anhui Science and Technology University, Fengyang 233100, Anhui, China
| | - Guijie Chen
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences & Technology, Anhui Agricultural University, Hefei 230036, Anhui, China
| | - Kun Yu
- College of Food Engineering, Anhui Science and Technology University, Fengyang 233100, Anhui, China
| | - Chuanlai Du
- College of Food Engineering, Anhui Science and Technology University, Fengyang 233100, Anhui, China
| | - Wenbiao Shen
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Yi Sun
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Xiaoxiong Zeng
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| |
Collapse
|
28
|
Kuntz S, Kunz C, Borsch C, Hill D, Morrin S, Buck R, Rudloff S. Influence of microbially fermented 2´-fucosyllactose on neuronal-like cell activity in an in vitro co-culture system. Front Nutr 2024; 11:1351433. [PMID: 38389793 PMCID: PMC10881714 DOI: 10.3389/fnut.2024.1351433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 01/18/2024] [Indexed: 02/24/2024] Open
Abstract
Scope 2´-Fucosyllactose (2´-FL), the most abundant oligosaccharide in human milk, plays an important role in numerous biological functions, including improved learning. It is not clear, however, whether 2´-FL or a cleavage product could influence neuronal cell activity. Thus, we investigated the effects of 2´-FL, its monosaccharide fucose (Fuc), and microbial fermented 2´-FL and Fuc on the parameters of neuronal cell activity in an intestinal-neuronal transwell co-culture system in vitro. Methods Native 13C-labeled 2´-FL and 13C-Fuc or their metabolites, fermented with Bifidobacterium (B.) longum ssp. infantis and B. breve, which were taken from the lag-, log- and stationary (stat-) growth phases of batch cultures, were applied to the apical compartment of the co-culture system with Caco-2 cells representing the intestinal layer and all-trans-retinoic acid-differentiated SH-SY5Y (SH-SY5YATRA) cells mimicking neuronal-like cells. After 3 h of incubation, the culture medium in the basal compartment was monitored for 13C enrichment by using elemental analysis isotope-ratio mass spectrometry (EA-IRMS) and effects on cell viability, plasma, and mitochondrial membrane potential. The neurotransmitter activation (BDNF, GABA, choline, and glutamate) of SH-SY5YATRA cells was also determined. Furthermore, these effects were also measured by the direct application of 13C-2´-FL and 13C-Fuc to SH-SY5YATRA cells. Results While no effects on neuronal-like cell activities were observed after intact 2´-FL or Fuc was incubated with SH-SY5YATRA cells, supernatants from the stat-growth phase of 2´-FL, fermented by B. longum ssp. infantis alone and together with B. breve, significantly induced BDNF release from SH-SY5YATRA cells. No such effects were found for 2´-FL, Fuc, or their fermentation products from B. breve. The BDNF release occurred from an enhanced vesicular release, which was confirmed by the use of the Ca2+-channel blocker verapamil. Concomitant with this event, 13C enrichment was also observed in the basal compartment when supernatants from the stat-growth phase of fermentation by B. longum ssp. infantis alone or together with B. breve were used. Conclusion The results obtained in this study suggest that microbial products of 2´-FL rather than the oligosaccharide itself may influence neuronal cell activities.
Collapse
Affiliation(s)
- Sabine Kuntz
- Department of Nutritional Science, Justus Liebig University Giessen, Giessen, Germany
| | - Clemens Kunz
- Department of Nutritional Science, Justus Liebig University Giessen, Giessen, Germany
| | - Christian Borsch
- Department of Nutritional Science, Justus Liebig University Giessen, Giessen, Germany
| | - David Hill
- Abbott, Nutrition Division, Columbus, OH, United States
| | - Sinéad Morrin
- Abbott, Nutrition Division, Columbus, OH, United States
| | - Rachael Buck
- Abbott, Nutrition Division, Columbus, OH, United States
| | - Silvia Rudloff
- Department of Nutritional Science, Justus Liebig University Giessen, Giessen, Germany
- Department of Pediatrics, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
29
|
Vaz SR, Tofoli MH, Avelino MAG, da Costa PSS. Probiotics for infantile colic: Is there evidence beyond doubt? A meta-analysis and systematic review. Acta Paediatr 2024; 113:170-182. [PMID: 37962097 DOI: 10.1111/apa.17036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/27/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023]
Abstract
AIM This study is a systematic review and meta-analysis of randomised controlled trials that employed probiotics and symbiotics for treating infantile colic. METHODS We performed electronic systematic literature searches in Embase, PubMed and Web of Science, to identify articles published between 1950 and April 2023. Only RCT involving infants with infantile colic under 3 months were included. The treatment plan comprised 15 probiotics, which included Lactobacillus reuteri DSM 17938 and Bifidobacterium animalis lactis BB-12. The probiotics were administered alone or in combination with a prebiotic, vs. no intervention or a placebo. RESULTS Probiotics resulted in an average reduction of 51 min of crying per day (p = 0.001). Further analysis of subgroups showed that the reduction was -39.30 min for vaginal delivery (p = 0.003), -64.66 min for Lactobacillus reuteri DSM 17938 (p = 0.03), -40.45 min for other strains (p < 0.00001), -74.28 min for exclusively breastfed infants (p = 0.0003) and -48.04 min for mixed feeding (p < 0.00001). CONCLUSION All probiotic strains seem effective in treating infantile colic. Exclusively breastfed infants have demonstrated more significant reduction in crying time. However, the available evidence on the effectiveness of probiotics in formula-fed and caesarean-born infants is limited.
Collapse
Affiliation(s)
| | - Marise Helena Tofoli
- Department of Pediatric Gastroenterology, State Hospital for Children and Adolescents, Goiânia, Brazil
| | | | | |
Collapse
|
30
|
Huang X, Liu R, Wang J, Bao Y, Yi H, Wang X, Lu Y. Preparation and synbiotic interaction mechanism of microcapsules of Bifidobacterium animalis F1-7 and human milk oligosaccharides (HMO). Int J Biol Macromol 2024; 259:129152. [PMID: 38176500 DOI: 10.1016/j.ijbiomac.2023.129152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 12/21/2023] [Accepted: 12/28/2023] [Indexed: 01/06/2024]
Abstract
Probiotics such as Bifidobacterium spp. generally possess important physiological functions. However, maintaining probiotic viability is a challenge during processing, storage, and digestive transit period. Microencapsulation is widely considered to be an attractive approach. In this study, B. animalis F1-7 microcapsules and B. animalis F1-7-HMO microcapsules were successfully prepared by emulsification/internal gelation with high encapsulation efficiency (90.67 % and 92.16 %, respectively). The current study revealed that HMO-supplemented microcapsules exhibited more stable lyophilized forms and thermal stability. Additionally, a significant improvement in probiotic cell viability was observed in such microcapsules during simulated gastrointestinal (GI) fluids or storage. We also showed that the individual HMO mixtures 6'-SL remarkably promoted the growth and acetate yield of B. animalis F1-7 for 48 h (p < 0.05). The synbiotic combination of 6'-SL with B. animalis F1-7 enhanced SCFAs production in vitro fecal fermentation, decreasing several harmful intestinal bacteria such as Dorea, Escherichia-Shigella, and Streptococcus while enriching the probiotic A. muciniphila. This study provides strong support for HMO or 6'-SL combined with B. animalis F1-7 as an innovative dietary ingredient to bring health benefits. The potential of the synbiotic microcapsules with this combination merits further exploration for future use in the food industry.
Collapse
Affiliation(s)
- Xiaoyang Huang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Rui Liu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Jing Wang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Yuexin Bao
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Huaxi Yi
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, Shandong, China
| | - Xiaohong Wang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China; Key Laboratory of Environment Correlative Dietology, Ministry of Education (Huazhong Agricultural University), Wuhan 430070, Hubei, China
| | - Youyou Lu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China; Key Laboratory of Environment Correlative Dietology, Ministry of Education (Huazhong Agricultural University), Wuhan 430070, Hubei, China.
| |
Collapse
|
31
|
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
|
32
|
Fang X, Zhu W, Zhao H, Yin W, Song X, Liu F. Preclinical Safety Evaluation of the Human-Identical Milk Oligosaccharide Lacto-N-Triose II. Int J Toxicol 2024; 43:27-45. [PMID: 37769680 DOI: 10.1177/10915818231203515] [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: 10/03/2023]
Abstract
Lacto-N-triose II (LNT II), an essential human milk oligosaccharide and precursor to lacto-N-tetraose (LNT) and lacto-N-neotetraose (LNnT), was evaluated for safety. Genotoxicity was assessed through in vitro tests including Bacterial Reverse Mutation Test and mammalian cell micronucleus test, and a subchronic oral gavage toxicity study was conducted on juvenile Sprague-Dawley rats. In this study, LNT II was administered at dose levels of 0, 1,500, 2,500, or 5,000 mg/kg body weight (bw)/day for 90 days, followed by a 4-week treatment-free recovery period. LNT II was non-genotoxic in the in vitro assays. No compound-related effects were observed across all dosage levels based on various measures, including clinical observations, body weight gain, feed consumption, clinical pathology, organ weights, and histopathology. Consequently, the highest dosage of 5,000 mg/kg bw/day was established as the no-observed-adverse-effect-level (NOAEL). These results suggest the safe use of LNT II in young children formula and as a food ingredient, within the limits found naturally in human breast milk.
Collapse
Affiliation(s)
- Xu Fang
- Research and Development Department of Shandong Henglu Biotechnology Co., Ltd, Jinan, China
| | - Wei Zhu
- Technical Department, Anchor Center for Certification, Shanghai, China
| | - Huiying Zhao
- Specialty Toxicology Department, Pharmaron TSP, Beijing, China
| | - Wencheng Yin
- Research and Development Department of Shandong Henglu Biotechnology Co., Ltd, Jinan, China
| | - Xiao Song
- Research and Development Department of Shandong Henglu Biotechnology Co., Ltd, Jinan, China
| | - Fengxin Liu
- Research and Development Department of Shandong Henglu Biotechnology Co., Ltd, Jinan, China
| |
Collapse
|
33
|
Wang Y, Rui B, Ze X, Liu Y, Yu D, Liu Y, Li Z, Xi Y, Ning X, Lei Z, Yuan J, Li L, Zhang X, Li W, Deng Y, Yan J, Li M. Sialic acid-based probiotic intervention in lactating mothers improves the neonatal gut microbiota and immune responses by regulating sialylated milk oligosaccharide synthesis via the gut-breast axis. Gut Microbes 2024; 16:2334967. [PMID: 38630006 PMCID: PMC11028031 DOI: 10.1080/19490976.2024.2334967] [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: 07/05/2023] [Accepted: 03/21/2024] [Indexed: 04/19/2024] Open
Abstract
Human milk oligosaccharides (HMOs) are vital milk carbohydrates that help promote the microbiota-dependent growth and immunity of infants. Sialic acid (SA) is a crucial component of sialylated milk oligosaccharides (S-MOs); however, the effects of SA supplementation in lactating mothers on S-MO biosynthesis and their breastfed infants are unknown. Probiotic intervention during pregnancy or lactation demonstrates promise for modulating the milk glycobiome. Here, we evaluated whether SA and a probiotic (Pro) mixture could increase S-MO synthesis in lactating mothers and promote the microbiota development of their breastfed neonates. The results showed that SA+Pro intervention modulated the gut microbiota and 6'-SL contents in milk of maternal rats more than the SA intervention, which promoted Lactobacillus reuteri colonization in neonates and immune development. Deficient 6'-SL in the maternal rat milk of St6gal1 knockouts (St6gal1-/-) disturbed intestinal microbial structures in their offspring, thereby impeding immune tolerance development. SA+Pro intervention in lactating St6gal1± rats compromised the allergic responses of neonates by promoting 6'-SL synthesis and the neonatal gut microbiota. Our findings from human mammary epithelial cells (MCF-10A) indicated that the GPR41-PI3K-Akt-PPAR pathway helped regulate 6'-SL synthesis in mammary glands after SA+Pro intervention through the gut - breast axis. We further validated our findings using a human-cohort study, confirming that providing SA+Pro to lactating Chinese mothers increased S-MO contents in their breast milk and promoted gut Bifidobacterium spp. and Lactobacillus spp. colonization in infants, which may help enhance immune responses. Collectively, our findings may help alter the routine supplementation practices of lactating mothers to modulate milk HMOs and promote the development of early-life gut microbiota and immunity.
Collapse
Affiliation(s)
- Yushuang Wang
- Department of Microecology, College of Basic Medical Science, Dalian Medical University, Dalian, China
- Department of Clinical Laboratory, Central Hospital of Dalian University of Technology, Dalian, China
| | - Binqi Rui
- Department of Microecology, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Xiaolei Ze
- Microbiome Research and Application Center, BYHEALTH Institute of Nutrition & Health, Guangzhou, China
| | - Yujia Liu
- Department of Microecology, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Da Yu
- The Third Ward of Obstetrics and Gynecology at Chunliu District, Dalian Women and Children Medical Center (Group), Dalian, China
| | - Yinhui Liu
- Department of Microecology, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Zhi Li
- Department of Clinical Laboratory, Central Hospital of Dalian University of Technology, Dalian, China
| | - Yu Xi
- Microbiome Research and Application Center, BYHEALTH Institute of Nutrition & Health, Guangzhou, China
| | - Xixi Ning
- Department of Microecology, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Zengjie Lei
- Department of Microecology, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Jieli Yuan
- Department of Microecology, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Liang Li
- Microbiome Research and Application Center, BYHEALTH Institute of Nutrition & Health, Guangzhou, China
| | - Xuguang Zhang
- Microbiome Research and Application Center, BYHEALTH Institute of Nutrition & Health, Guangzhou, China
| | - Wenzhe Li
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| | - Yanjie Deng
- The Third Ward of Obstetrics and Gynecology at Chunliu District, Dalian Women and Children Medical Center (Group), Dalian, China
| | - Jingyu Yan
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences Key Laboratory of Separation Science for Analytical Chemistry, Dalian, China
| | - Ming Li
- Department of Microecology, College of Basic Medical Science, Dalian Medical University, Dalian, China
| |
Collapse
|
34
|
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
|
35
|
Zuurveld M, Diks MAP, Kiliaan PCJ, Garssen J, Folkerts G, van’t Land B, Willemsen LEM. Butyrate interacts with the effects of 2'FL and 3FL to modulate in vitro ovalbumin-induced immune activation, and 2'FL lowers mucosal mast cell activation in a preclinical model for hen's egg allergy. Front Nutr 2023; 10:1305833. [PMID: 38174112 PMCID: PMC10762782 DOI: 10.3389/fnut.2023.1305833] [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: 10/02/2023] [Accepted: 11/15/2023] [Indexed: 01/05/2024] Open
Abstract
Background Early life provides a window of opportunity to prevent allergic diseases. With a prevalence of 0.5-2% in infants, hen's egg allergy is one of the most common food allergies. The immunomodulatory effects of human milk oligosaccharides (HMOs), 2'-fucosyllactose (2'FL), and 3-fucosyllactose (3FL) were studied in an in vitro mucosal immune model and an in vivo murine model for hen's egg (ovalbumin) allergy. Methods Intestinal epithelial cell (IEC)/dendritic cell (DC) and DC/T cell cocultures were used to expose IECs to ovalbumin (OVA) in an in vitro mucosal immune model. The effects of epithelial pre-incubation with 0.1% 2'FL or 3FL and/or 0.5 mM butyrate were studied. Three- to four-weeks-old female C3H/HeOuJ mice were fed AIN93G diets containing 0.1-0.5% 2'FL or 3FL 2 weeks before and during OVA sensitization and challenge. Allergic symptoms and systemic and local immune parameters were assessed. Results Exposing IECs to butyrate in vitro left the IEC/DC/T cell cross-talk unaffected, while 2'FL and 3FL showed differential immunomodulatory effects. In 3FL exposed IEC-DC-T cells, the secretion of IFNγ and IL10 was enhanced. This was observed upon pre-incubation of IECs with 2'FL and butyrate as well, but not 2'FL alone. The presence of butyrate did not affect OVA activation, but when combined with 3FL, an increase in IL6 release from DCs was observed (p < 0.001). OVA allergic mice receiving 0.5% 3FL diet had a lower %Th2 cells in MLNs, but the humoral response was unaltered compared to control mice. OVA-allergic mice receiving 0.1 or 0.5% 2'FL diets had lower serum levels of OVA-IgG2a (p < 0.05) or the mast cell marker mMCP1, in association with increased concentration of cecal short-chain fatty acids (SCFAs) (p < 0.05). Conclusion In vitro butyrate exposure promotes the development of a downstream type 1 and regulatory response observed after 2'FL exposure. 2'FL and 3FL differentially modulate ovalbumin-induced mucosal inflammation predominantly independent of butyrate. Mice receiving dietary 3FL during ovalbumin sensitization and challenge had lowered Th2 activation while the frequency of Treg cells was enhanced. By contrast, 2'FL improved the humoral immune response and suppressed mast cell activation in association with increased SCFAs production in the murine model for hen's egg allergy.
Collapse
Affiliation(s)
- M. Zuurveld
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - M. A. P. Diks
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - P. C. J. Kiliaan
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - J. Garssen
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
- Danone Nutricia Research B.V, Utrecht, Netherlands
| | - G. Folkerts
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - B. van’t Land
- Danone Nutricia Research B.V, Utrecht, Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - L. E. M. Willemsen
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
36
|
Low KE, Tingley JP, Klassen L, King ML, Xing X, Watt C, Hoover SER, Gorzelak M, Abbott DW. Carbohydrate flow through agricultural ecosystems: Implications for synthesis and microbial conversion of carbohydrates. Biotechnol Adv 2023; 69:108245. [PMID: 37652144 DOI: 10.1016/j.biotechadv.2023.108245] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/10/2023] [Accepted: 08/25/2023] [Indexed: 09/02/2023]
Abstract
Carbohydrates are chemically and structurally diverse biomolecules, serving numerous and varied roles in agricultural ecosystems. Crops and horticulture products are inherent sources of carbohydrates that are consumed by humans and non-human animals alike; however carbohydrates are also present in other agricultural materials, such as soil and compost, human and animal tissues, milk and dairy products, and honey. The biosynthesis, modification, and flow of carbohydrates within and between agricultural ecosystems is intimately related with microbial communities that colonize and thrive within these environments. Recent advances in -omics techniques have ushered in a new era for microbial ecology by illuminating the functional potential for carbohydrate metabolism encoded within microbial genomes, while agricultural glycomics is providing fresh perspective on carbohydrate-microbe interactions and how they influence the flow of functionalized carbon. Indeed, carbohydrates and carbohydrate-active enzymes are interventions with unrealized potential for improving carbon sequestration, soil fertility and stability, developing alternatives to antimicrobials, and circular production systems. In this manner, glycomics represents a new frontier for carbohydrate-based biotechnological solutions for agricultural systems facing escalating challenges, such as the changing climate.
Collapse
Affiliation(s)
- Kristin E Low
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, Lethbridge, AB, Canada
| | - Jeffrey P Tingley
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, Lethbridge, AB, Canada
| | - Leeann Klassen
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, Lethbridge, AB, Canada
| | - Marissa L King
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, Lethbridge, AB, Canada
| | - Xiaohui Xing
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, Lethbridge, AB, Canada
| | - Caitlin Watt
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, Lethbridge, AB, Canada
| | - Shelley E R Hoover
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB, Canada
| | - Monika Gorzelak
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, Lethbridge, AB, Canada
| | - D Wade Abbott
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, Lethbridge, AB, Canada.
| |
Collapse
|
37
|
Kiely LJ, Busca K, Lane JA, van Sinderen D, Hickey RM. Molecular strategies for the utilisation of human milk oligosaccharides by infant gut-associated bacteria. FEMS Microbiol Rev 2023; 47:fuad056. [PMID: 37793834 PMCID: PMC10629584 DOI: 10.1093/femsre/fuad056] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 09/14/2023] [Accepted: 10/03/2023] [Indexed: 10/06/2023] Open
Abstract
A number of bacterial species are found in high abundance in the faeces of healthy breast-fed infants, an occurrence that is understood to be, at least in part, due to the ability of these bacteria to metabolize human milk oligosaccharides (HMOs). HMOs are the third most abundant component of human milk after lactose and lipids, and represent complex sugars which possess unique structural diversity and are resistant to infant gastrointestinal digestion. Thus, these sugars reach the infant distal intestine intact, thereby serving as a fermentable substrate for specific intestinal microbes, including Firmicutes, Proteobacteria, and especially infant-associated Bifidobacterium spp. which help to shape the infant gut microbiome. Bacteria utilising HMOs are equipped with genes associated with their degradation and a number of carbohydrate-active enzymes known as glycoside hydrolase enzymes have been identified in the infant gut, which supports this hypothesis. The resulting degraded HMOs can also be used as growth substrates for other infant gut bacteria present in a microbe-microbe interaction known as 'cross-feeding'. This review describes the current knowledge on HMO metabolism by particular infant gut-associated bacteria, many of which are currently used as commercial probiotics, including the distinct strategies employed by individual species for HMO utilisation.
Collapse
Affiliation(s)
- Leonie Jane Kiely
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork P61C996, Ireland
- Health and Happiness Group, H&H Research, National Food Innovation Hub, Teagasc Moorepark, Fermoy, Co. Cork P61K202, Ireland
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12 YT20, Ireland
- School of Microbiology, University College Cork, Cork T12 YN60, Ireland
| | - Kizkitza Busca
- Health and Happiness Group, H&H Research, National Food Innovation Hub, Teagasc Moorepark, Fermoy, Co. Cork P61K202, Ireland
| | - Jonathan A Lane
- Health and Happiness Group, H&H Research, National Food Innovation Hub, Teagasc Moorepark, Fermoy, Co. Cork P61K202, Ireland
| | - Douwe van Sinderen
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12 YT20, Ireland
- School of Microbiology, University College Cork, Cork T12 YN60, Ireland
| | - Rita M Hickey
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork P61C996, Ireland
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12 YT20, Ireland
| |
Collapse
|
38
|
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
|
39
|
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
|
40
|
Barreiros-Mota I, R. Araújo J, Marques C, Sousa L, Morais J, Castela I, Faria A, Neto MT, Cordeiro-Ferreira G, Virella D, Pita A, Pereira-da-Silva L, Calhau C. Changes in Microbiota Profile in the Proximal Remnant Intestine in Infants Undergoing Surgery Requiring Enterostomy. Microorganisms 2023; 11:2482. [PMID: 37894140 PMCID: PMC10609405 DOI: 10.3390/microorganisms11102482] [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: 09/20/2023] [Revised: 09/29/2023] [Accepted: 09/30/2023] [Indexed: 10/29/2023] Open
Abstract
Early-life gut dysbiosis has been associated with an increased risk of inflammatory, metabolic, and immune diseases later in life. Data on gut microbiota changes in infants undergoing intestinal surgery requiring enterostomy are scarce. This prospective cohort study examined the enterostomy effluent of 29 infants who underwent intestinal surgery due to congenital malformations of the gastrointestinal tract, necrotizing enterocolitis, or spontaneous intestinal perforation. Initial effluent samples were collected immediately after surgery and final effluent samples were collected three weeks later. Gut microbiota composition was analysed using real-time PCR and 16S rRNA gene sequencing. Three weeks after surgery, an increase in total bacteria number (+21%, p = 0.026), a decrease in Staphylococcus (-21%, p = 0.002) and Candida spp. (-16%, p = 0.045), and an increase in Lactobacillus (+3%, p = 0.045) and in less abundant genera belonging to the Enterobacteriales family were found. An increase in alpha diversity (Shannon's and Simpson's indexes) and significant alterations in beta diversity were observed. A correlation of necrotizing enterocolitis with higher Staphylococcus abundance and higher alpha diversity was also observed. H2-blockers and/or proton pump inhibitor therapy were positively correlated with a higher total bacteria number. In conclusion, these results suggest that positive changes occur in the gut microbiota profile of infants three weeks after intestinal surgery.
Collapse
Affiliation(s)
- Inês Barreiros-Mota
- Nutrition & Metabolism Department, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal; (I.B.-M.); (J.R.A.); (C.M.); (L.S.); (I.C.); (A.F.)
- CHRC—Comprehensive Health Research Centre, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal; (J.M.); (M.T.N.); (L.P.-d.-S.)
| | - João R. Araújo
- Nutrition & Metabolism Department, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal; (I.B.-M.); (J.R.A.); (C.M.); (L.S.); (I.C.); (A.F.)
- Nutrition & Metabolism Department, CINTESIS@RISE, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal
| | - Cláudia Marques
- Nutrition & Metabolism Department, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal; (I.B.-M.); (J.R.A.); (C.M.); (L.S.); (I.C.); (A.F.)
- Nutrition & Metabolism Department, CINTESIS@RISE, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal
| | - Laura Sousa
- Nutrition & Metabolism Department, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal; (I.B.-M.); (J.R.A.); (C.M.); (L.S.); (I.C.); (A.F.)
| | - Juliana Morais
- CHRC—Comprehensive Health Research Centre, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal; (J.M.); (M.T.N.); (L.P.-d.-S.)
| | - Inês Castela
- Nutrition & Metabolism Department, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal; (I.B.-M.); (J.R.A.); (C.M.); (L.S.); (I.C.); (A.F.)
- CHRC—Comprehensive Health Research Centre, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal; (J.M.); (M.T.N.); (L.P.-d.-S.)
| | - Ana Faria
- Nutrition & Metabolism Department, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal; (I.B.-M.); (J.R.A.); (C.M.); (L.S.); (I.C.); (A.F.)
- CHRC—Comprehensive Health Research Centre, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal; (J.M.); (M.T.N.); (L.P.-d.-S.)
- Nutrition & Metabolism Department, CINTESIS@RISE, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal
| | - Maria Teresa Neto
- CHRC—Comprehensive Health Research Centre, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal; (J.M.); (M.T.N.); (L.P.-d.-S.)
- Neonatal Intensive Care Unit, Department of Pediatrics, Hospital Dona Estefânia, Centro Hospitalar Universitário de Lisboa Central, 1169-045 Lisbon, Portugal; (G.C.-F.); (D.V.); (A.P.)
- Medicine of Woman, Childhood and Adolescence Academic Area, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), 1169-056 Lisbon, Portugal
| | - Gonçalo Cordeiro-Ferreira
- Neonatal Intensive Care Unit, Department of Pediatrics, Hospital Dona Estefânia, Centro Hospitalar Universitário de Lisboa Central, 1169-045 Lisbon, Portugal; (G.C.-F.); (D.V.); (A.P.)
| | - Daniel Virella
- Neonatal Intensive Care Unit, Department of Pediatrics, Hospital Dona Estefânia, Centro Hospitalar Universitário de Lisboa Central, 1169-045 Lisbon, Portugal; (G.C.-F.); (D.V.); (A.P.)
| | - Ana Pita
- Neonatal Intensive Care Unit, Department of Pediatrics, Hospital Dona Estefânia, Centro Hospitalar Universitário de Lisboa Central, 1169-045 Lisbon, Portugal; (G.C.-F.); (D.V.); (A.P.)
| | - Luís Pereira-da-Silva
- CHRC—Comprehensive Health Research Centre, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal; (J.M.); (M.T.N.); (L.P.-d.-S.)
- Neonatal Intensive Care Unit, Department of Pediatrics, Hospital Dona Estefânia, Centro Hospitalar Universitário de Lisboa Central, 1169-045 Lisbon, Portugal; (G.C.-F.); (D.V.); (A.P.)
- Medicine of Woman, Childhood and Adolescence Academic Area, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), 1169-056 Lisbon, Portugal
| | - Conceição Calhau
- Nutrition & Metabolism Department, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal; (I.B.-M.); (J.R.A.); (C.M.); (L.S.); (I.C.); (A.F.)
- Nutrition & Metabolism Department, CINTESIS@RISE, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal
| |
Collapse
|
41
|
Li C, Li M, Hu M, Zhang T. Metabolic Engineering of De Novo Pathway for the Production of 2'-Fucosyllactose in Escherichia coli. Mol Biotechnol 2023; 65:1485-1497. [PMID: 36652181 DOI: 10.1007/s12033-023-00657-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 01/05/2023] [Indexed: 01/19/2023]
Abstract
2'-Fucosyllactose (2'-FL), one of the most abundant oligosaccharides in human milk, has gained increased attention owing to its nutraceutical and pharmaceutical potential. However, limited availability and high-cost of preparation have limited its widespread application and in-depth investigation of its potential functions. Here, a modular pathway engineering was implemented to construct an Escherichia coli strain to improve the biosynthesis titer of 2'-FL. Before overexpression of manB, manC, gmd, wcaG, and heterologous expression of futC, genes wcaJ and lacZ encoding UDP-glucose lipid carrier transferase and β-galactosidase, respectively, were inactivated from E. coli BL21 (DE3) with the CRISPR-Cas9 system, which inhibited the production of 2'-FL. The results showed that final shake flask culture yielded a 3.8-fold increase in 2'-FL (0.98 g/L) from the engineered strain ELC07. Fed-batch fermentation conditions were optimized in a 3-L bioreactor. The highest titer of 2'-FL (18.22 g/L) was obtained, corresponding to a yield of 0.25 g/g glycerol and a substrate conversion of 0.88 g/g lactose.
Collapse
Affiliation(s)
- Chenchen Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Mengli Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Miaomiao Hu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Tao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China.
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, 214122, Jiangsu, China.
| |
Collapse
|
42
|
Mollova D, Vasileva T, Bivolarski V, Iliev I. The Enzymatic Hydrolysis of Human Milk Oligosaccharides and Prebiotic Sugars from LAB Isolated from Breast Milk. Microorganisms 2023; 11:1904. [PMID: 37630464 PMCID: PMC10458952 DOI: 10.3390/microorganisms11081904] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
Breastfeeding is essential in the first months of a newborn's life. Breast milk is a source of crucial macronutrients, prebiotic oligosaccharides, and potential probiotic strains of bacteria. Oligosaccharides from breast milk (HMOs) are a significant part of the composition of breast milk and represent a complex of digestible sugars. This study aims to elucidate the enzymatic hydrolysis of these oligosaccharides and other prebiotics by the bacteria present in breast milk. We used modified methods to isolate oligosaccharides (HMOs) from human milk. Using unique techniques, we isolated and identified different bacteria from breast milk, mainly Lactobacillus fermentum. Using enzymatic analyses, we established the participation of α-fucosidase, α-glucosidase, β-galactosidase, and β-glucosidase from breast milk bacteria in the hydrolysis of prebiotic sugars. We also optimized the scheme for isolating oligosaccharides from breast milk by putting the lyophilized product into different food media. We found that the oligosaccharides from breast milk (HMOs) are a potent inducer for the secretion of the studied bacterial enzymes. Also, we found that all the lactobacilli strains we studied in detail could digest mucin-linked glycans. The degradation of these sugars is perhaps a built-in defense mechanism in cases where other sugars are lacking in the environment. We also determined fucosidase activity in some of the isolated strains. We recorded the highest values (2.5 U/mg in L. fermentum ss8) when the medium's oligosaccharides isolated from breast milk were present. Lactobacilli and Bifidobacteria supplied with breast milk are the first colonizers in most cases in the gastrointestinal tract of the newborn. The presence and study of different genes for synthesizing other enzyme systems and transporters of various sugars in this type of bacteria are essential.
Collapse
Affiliation(s)
- Daniela Mollova
- Department of Biochemistry and Microbiology, Faculty of Biology, Plovdiv University “Paisii Hilendarski”, 4000 Plovdiv, Bulgaria; (T.V.); (V.B.); (I.I.)
| | | | | | | |
Collapse
|
43
|
Wijenayake S, Martz J, Lapp HE, Storm JA, Champagne FA, Kentner AC. The contributions of parental lactation on offspring development: It's not udder nonsense! Horm Behav 2023; 153:105375. [PMID: 37269591 PMCID: PMC10351876 DOI: 10.1016/j.yhbeh.2023.105375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 05/11/2023] [Accepted: 05/13/2023] [Indexed: 06/05/2023]
Abstract
The Developmental Origins of Health and Disease (DOHaD) hypothesis describes how maternal stress exposures experienced during critical periods of perinatal life are linked to altered developmental trajectories in offspring. Perinatal stress also induces changes in lactogenesis, milk volume, maternal care, and the nutritive and non-nutritive components of milk, affecting short and long-term developmental outcomes in offspring. For instance, selective early life stressors shape the contents of milk, including macro/micronutrients, immune components, microbiota, enzymes, hormones, milk-derived extracellular vesicles, and milk microRNAs. In this review, we highlight the contributions of parental lactation to offspring development by examining changes in the composition of breast milk in response to three well-characterized maternal stressors: nutritive stress, immune stress, and psychological stress. We discuss recent findings in human, animal, and in vitro models, their clinical relevance, study limitations, and potential therapeutic significance to improving human health and infant survival. We also discuss the benefits of enrichment methods and support tools that can be used to improve milk quality and volume as well as related developmental outcomes in offspring. Lastly, we use evidence-based primary literature to convey that even though select maternal stressors may modulate lactation biology (by influencing milk composition) depending on the severity and length of exposure, exclusive and/or prolonged milk feeding may attenuate the negative in utero effects of early life stressors and promote healthy developmental trajectories. Overall, scientific evidence supports lactation to be protective against nutritive and immune stressors, but the benefits of lactation in response to psychological stressors need further investigation.
Collapse
Affiliation(s)
- Sanoji Wijenayake
- Department of Biology, The University of Winnipeg, Winnipeg, Manitoba, Canada.
| | - Julia Martz
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, USA
| | - Hannah E Lapp
- Deparment of Psychology, University of Texas at Austin, Austin, TX, USA
| | - Jasmyne A Storm
- Department of Biology, The University of Winnipeg, Winnipeg, Manitoba, Canada
| | | | - Amanda C Kentner
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, USA.
| |
Collapse
|
44
|
Salli K, Hirvonen J, Anglenius H, Hibberd AA, Ahonen I, Saarinen MT, Maukonen J, Ouwehand AC. The Effect of Human Milk Oligosaccharides and Bifidobacterium longum subspecies infantis Bi-26 on Simulated Infant Gut Microbiome and Metabolites. Microorganisms 2023; 11:1553. [PMID: 37375055 DOI: 10.3390/microorganisms11061553] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Human milk oligosaccharides (HMOs) shape the developing infant gut microbiota. In this study, a semi-continuous colon simulator was used to evaluate the effect of 2 HMOs-2'-fucosyllactose (2'-FL) and 3-fucosyllactose (3-FL)-on the composition of infant faecal microbiota and microbial metabolites. The simulations were performed with and without a probiotic Bifidobacterium longum subspecies infantis Bi-26 (Bi-26) and compared with a control that lacked an additional carbon source. The treatments with HMOs decreased α-diversity and increased Bifidobacterium species versus the control, but the Bifidobacterium species differed between simulations. The levels of acetic acid and the sum of all short-chain fatty acids (SCFAs) trended toward an increase with 2'-FL, as did lactic acid with 2'-FL and 3-FL, compared with control. A clear correlation was seen between the consumption of HMOs and the increase in SCFAs (-0.72) and SCFAs + lactic acid (-0.77), whereas the correlation between HMO consumption and higher total bifidobacterial numbers was moderate (-0.46). Bi-26 decreased propionic acid levels with 2'-FL. In conclusion, whereas infant faecal microbiota varied between infant donors, the addition of 2'-FL and 3-FL, alone or in combination, increased the relative abundance and numbers Bifidobacterium species in the semi-continuous colon simulation model, correlating with the production of microbial metabolites. These findings may suggest that HMOs and probiotics benefit the developing infant gut microbiota.
Collapse
Affiliation(s)
- Krista Salli
- Global Health & Nutrition Science, IFF Health, 02460 Kantvik, Finland
| | - Johanna Hirvonen
- Global Health & Nutrition Science, IFF Health, 02460 Kantvik, Finland
| | - Heli Anglenius
- Global Health & Nutrition Science, IFF Health, 02460 Kantvik, Finland
| | - Ashley A Hibberd
- Genomics & Microbiome Science, IFF Health, Madison, WI 53716, USA
| | | | - Markku T Saarinen
- Global Health & Nutrition Science, IFF Health, 02460 Kantvik, Finland
| | - Johanna Maukonen
- Global Health & Nutrition Science, IFF Health, 02460 Kantvik, Finland
| | - Arthur C Ouwehand
- Global Health & Nutrition Science, IFF Health, 02460 Kantvik, Finland
| |
Collapse
|
45
|
Hill DR, Buck RH. Infants Fed Breastmilk or 2'-FL Supplemented Formula Have Similar Systemic Levels of Microbiota-Derived Secondary Bile Acids. Nutrients 2023; 15:nu15102339. [PMID: 37242222 DOI: 10.3390/nu15102339] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/05/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Human milk represents an optimal source of nutrition during infancy. Milk also serves as a vehicle for the transfer of growth factors, commensal microbes, and prebiotic compounds to the immature gastrointestinal tract. These immunomodulatory and prebiotic functions of milk are increasingly appreciated as critical factors in the development of the infant gut and its associated microbial community. Advances in infant formula composition have sought to recapitulate some of the prebiotic and immunomodulatory functions of milk through human milk oligosaccharide (HMO) fortification, with the aim of promoting healthy development both within the gastrointestinal tract and systemically. Our objective was to investigate the effects of feeding formulas supplemented with the HMO 2'-fucosyllactose (2'-FL) on serum metabolite levels relative to breastfed infants. A prospective, randomized, double-blinded, controlled study of infant formulas (64.3 kcal/dL) fortified with varying levels of 2'-FL and galactooligosaccharides (GOS) was conducted [0.2 g/L 2'-FL + 2.2 g/L GOS; 1.0 g/L 2'-FL + 1.4 g/L GOS]. Healthy singleton infants age 0-5 days and with birth weight > 2490 g were enrolled (n = 201). Mothers chose to either exclusively formula-feed or breastfeed their infant from birth to 4 months of age. Blood samples were drawn from a subset of infants at 6 weeks of age (n = 35-40 per group). Plasma was evaluated by global metabolic profiling and compared to a breastfed reference group (HM) and a control formula (2.4 g/L GOS). Fortification of control infant formula with the HMO 2'-FL resulted in significant increases in serum metabolites derived from microbial activity in the gastrointestinal tract. Most notably, secondary bile acid production was broadly increased in a dose-dependent manner among infants receiving 2'-FL supplemented formula relative to the control formula. 2'-FL supplementation increased secondary bile acid production to levels associated with breastfeeding. Our data indicate that supplementation of infant formula with 2'-FL supports the production of secondary microbial metabolites at levels comparable to breastfed infants. Thus, dietary supplementation of HMO may have broad implications for the function of the gut microbiome in systemic metabolism. This trial was registered at with the U.S. National library of Medicine as NCT01808105.
Collapse
Affiliation(s)
- David R Hill
- Abbott, Nutrition Division, Columbus, OH 43219, USA
| | | |
Collapse
|
46
|
Hiraku A, Nakata S, Murata M, Xu C, Mutoh N, Arai S, Odamaki T, Iwabuchi N, Tanaka M, Tsuno T, Nakamura M. Early Probiotic Supplementation of Healthy Term Infants with Bifidobacterium longum subsp. infantis M-63 Is Safe and Leads to the Development of Bifidobacterium-Predominant Gut Microbiota: A Double-Blind, Placebo-Controlled Trial. Nutrients 2023; 15:1402. [PMID: 36986131 PMCID: PMC10055625 DOI: 10.3390/nu15061402] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 02/25/2023] [Accepted: 03/02/2023] [Indexed: 03/17/2023] Open
Abstract
Bifidobacteria are important intestinal bacteria that provide a variety of health benefits in infants. We investigated the efficacy and safety of Bifidobacterium longum subsp. infantis (B. infantis) M-63 in healthy infants in a double-blind, randomized, placebo-controlled trial. Healthy term infants were given B. infantis M-63 (n = 56; 1 × 109 CFU/day) or placebo (n = 54) from postnatal age ≤ 7 days to 3 months. Fecal samples were collected, and fecal microbiota, stool pH, short-chain fatty acids, and immune substances were analyzed. Supplementation with B. infantis M-63 significantly increased the relative abundance of Bifidobacterium compared with the placebo group, with a positive correlation with the frequency of breastfeeding. Supplementation with B. infantis M-63 led to decreased stool pH and increased levels of acetic acid and IgA in the stool at 1 month of age compared with the placebo group. There was a decreased frequency of defecation and watery stools in the probiotic group. No adverse events related to test foods were observed. These results indicate that early supplementation with B. infantis M-63 is well tolerated and contributes to the development of Bifidobacterium-predominant gut microbiota during a critical developmental phase in term infants.
Collapse
Affiliation(s)
- Akari Hiraku
- Food Ingredients and Technology Institute, R & D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama 252-8583, Japan
| | - Setsuko Nakata
- Department of Pediatrics, Matsumoto City Hospital, 4417-180, Hata, Matsumoto 390-1401, Japan
| | - Mai Murata
- Food Ingredients and Technology Institute, R & D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama 252-8583, Japan
| | - Chendong Xu
- Food Ingredients and Technology Institute, R & D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama 252-8583, Japan
| | - Natsumi Mutoh
- Food Ingredients and Technology Institute, R & D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama 252-8583, Japan
| | - Satoshi Arai
- Food Ingredients and Technology Institute, R & D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama 252-8583, Japan
| | - Toshitaka Odamaki
- Next Generation Science Institute, R & D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama 252-8583, Japan
| | - Noriyuki Iwabuchi
- Food Ingredients and Technology Institute, R & D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama 252-8583, Japan
| | - Miyuki Tanaka
- Food Ingredients and Technology Institute, R & D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama 252-8583, Japan
| | - Takahisa Tsuno
- Department of Pediatrics, Matsumoto City Hospital, 4417-180, Hata, Matsumoto 390-1401, Japan
| | - Masahiko Nakamura
- Department of neurosurgery, Matsumoto City Hospital, 4417-180, Hata, Matsumoto 390-1401, Japan
| |
Collapse
|
47
|
Zhang G, Sun H, Xu Z, Tan Z, Xiao L, He M, Shang J, Tsapieva AN, Zhang L. Screening of Bifidobacteria with Probiotic Potential from Healthy Infant Feces by Using 2'-Fucosyllactose. Foods 2023; 12:foods12040858. [PMID: 36832933 PMCID: PMC9957139 DOI: 10.3390/foods12040858] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/14/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023] Open
Abstract
Using 2'-fucosyllactose (2'-FL) as the sole carbon source can be an efficient way to screen bifidobacteria with superior probiotic capabilities since 2'-FL is a key element in promoting the growth of intestinal bifidobacteria in newborns. This approach was used in this work to screen eight bifidobacteria strains, including one strain of Bifidobacterium longum subsp. infantis BI_Y46 and seven strains of Bifidobacterium bifidum (BB_Y10, BB_Y30, BB_Y39, BB_S40, BB_H4, BB_H5 and BB_H22). Studies on their probiotic properties showed that BI_Y46 had a unique morphology with pilus-like structure, a high resistance to bile salt stimulation and a potent inhibitory action on Escherichia coli ATCC 25922. Similarly, BB_H5 and BB_H22 produced more extracellular polysaccharides and had a higher protein content than other strains. In contrast, BB_Y22 displayed considerable auto-aggregation activity and a high resistance to bile salt stimulation. Interestingly, BB_Y39 with weak self-aggregation ability and acid resistance had very excellent bile salt tolerance, extracellular polysaccharides (EPS) production and bacteriostatic ability. In conclusion, 2'-FL was used as sole carbon source to identify eight bifidobacteria with excellent probiotic properties.
Collapse
Affiliation(s)
- Gongsheng Zhang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Hui Sun
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Zihe Xu
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Ze Tan
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Lihong Xiao
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Mingxue He
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Jiaqi Shang
- Key Laboratory of Bionic Engineering, Ministry of Education, College of Biological and Agricultural Engineering, Jilin University, Changchun 130022, China
| | - Anna N. Tsapieva
- Department of Molecular Microbiology, FSBSI Institute of Experimental Medicine, Acad. Pavlov Street, 12, 197376 St. Petersburg, Russia
| | - Lili Zhang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
- Correspondence: ; Tel.: +86-451-5519-0675
| |
Collapse
|
48
|
Li R, Zhou Y, Xu Y. Comparative Analysis of Oligosaccharides in Breast Milk and Feces of Breast-Fed Infants by Using LC-QE-HF-MS: A Communication. Nutrients 2023; 15:nu15040888. [PMID: 36839244 PMCID: PMC9963387 DOI: 10.3390/nu15040888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
Currently, it must be acknowledged that little is known about the quantity and make-up of oligosaccharides (OS) found in breast-fed babies' feces as well as their metabolic fate. In the present work, UPLC-QE-HF-MS was successfully adopted to identify the profiles of human milk oligosaccharides (HMOs) in the breast milk of four mothers and fecal OS in the feces of their breast-fed infant. There were significant variations and differences in both number and composition between HMOs and fecal OS. The early-life gastrointestinal microbiota metabolism may be triggered into the advanced breakdown, synthesis, bioconversion, or redesign of HMOs. The fate of HMOs during passage through the gastrointestinal tract may be profoundly informed by the comparison of OS between breast milk and fecal OS profiles. The characterization of fecal OS could be applied as a valuable tool for monitoring the gastrointestinal fate of HMOs and reflecting infant development at different stages of lactation. Further research on the gastrointestinal bioconversion of HMOs profiles is required, including secretor type and the lactation time of milk, as well as baby feeding.
Collapse
Affiliation(s)
- Rui Li
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, No. 38 Xueyuan Road, Beijing 100083, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, No. 38 Xueyuan Road, Beijing 100083, China
- PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, No. 38 Xueyuan Road, Beijing 100083, China
| | - Yalin Zhou
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, No. 38 Xueyuan Road, Beijing 100083, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, No. 38 Xueyuan Road, Beijing 100083, China
| | - Yajun Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, No. 38 Xueyuan Road, Beijing 100083, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, No. 38 Xueyuan Road, Beijing 100083, China
- PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, No. 38 Xueyuan Road, Beijing 100083, China
- Correspondence: ; Tel.: +86-010-8280-2552
| |
Collapse
|
49
|
Zhu Y, Zhang J, Zhang W, Mu W. Recent progress on health effects and biosynthesis of two key sialylated human milk oligosaccharides, 3'-sialyllactose and 6'-sialyllactose. Biotechnol Adv 2023; 62:108058. [PMID: 36372185 DOI: 10.1016/j.biotechadv.2022.108058] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 10/25/2022] [Accepted: 11/06/2022] [Indexed: 11/13/2022]
Abstract
Human milk oligosaccharides (HMOs), the third major solid component in breast milk, are recognized as the first prebiotics for health benefits in infants. Sialylated HMOs are an important type of HMOs, accounting for approximately 13% of total HMOs. 3'-Sialyllactose (3'-SL) and 6'-sialyllactose (6'-SL) are two simplest sialylated HMOs. Both SLs display promising prebiotic effects, especially in promoting the proliferation of bifidobacteria and shaping the gut microbiota. SLs exhibit several health effects, including antiadhesive antimicrobial ability, antiviral activity, prevention of necrotizing enterocolitis, immunomodulatory activity, regulation of intestinal epithelial cell response, promotion of brain development, and cognition improvement. Both SLs have been approved as "Generally Recognized as Safe" by the American Food and Drug Administration and are commercially added to infant formula. The biosynthesis of SLs using enzymatic or microbial approaches has been widely studied. The enzymatic synthesis of SLs can be realized by two types of enzymes, sialidases with trans-sialidase activity and sialyltransferases. Microbial synthesis can be achieved by the multiple recombinant bacteria in one-pot reaction, which express the enzymes involved in SL synthesis pathways separately or in combination, or by metabolically engineered strains in a fermentation process. In this article, the physiological properties of 3'-SL and 6'-SL are summarized in detail and the biosynthesis of these SLs via enzymatic and microbial synthesis is comprehensively reviewed.
Collapse
Affiliation(s)
- Yingying Zhu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jiameng Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wenli Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wanmeng Mu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
50
|
DeVeaux A, Ryou J, Dantas G, Warner BB, Tarr PI. Microbiome-targeting therapies in the neonatal intensive care unit: safety and efficacy. Gut Microbes 2023; 15:2221758. [PMID: 37358104 PMCID: PMC10294772 DOI: 10.1080/19490976.2023.2221758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 05/25/2023] [Indexed: 06/27/2023] Open
Abstract
Microbiome-targeting therapies have received great attention as approaches to prevent disease in infants born preterm, but their safety and efficacy remain uncertain. Here we summarize the existing literature, focusing on recent meta-analyses and systematic reviews that evaluate the performance of probiotics, prebiotics, and/or synbiotics in clinical trials and studies, emphasizing interventions for which the primary or secondary outcomes were prevention of necrotizing enterocolitis, late-onset sepsis, feeding intolerance, and/or reduction in hospitalization length or all-cause mortality. Current evidence suggests that probiotics and prebiotics are largely safe but conclusions regarding their effectiveness in the neonatal intensive care unit have been mixed. To address this ambiguity, we evaluated publications that collectively support benefits of probiotics with moderate to high certainty evidence in a recent comprehensive network meta-analysis, highlighting limitations in these trials that make it difficult to support with confidence the routine, universal administration of probiotics to preterm infants.
Collapse
Affiliation(s)
- Anna DeVeaux
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jian Ryou
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Gautam Dantas
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Barbara B. Warner
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Phillip I. Tarr
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|