1
|
Naspolini NF, Schüroff PA, Vanzele PAR, Pereira-Santos D, Valim TA, Bonham KS, Fujita A, Passos-Bueno MR, Beltrão-Braga PCB, Carvalho ACPLF, Klepac-Ceraj V, Polanczyk GV, Campos AC, Taddei CR. Exclusive breastfeeding is associated with the gut microbiome maturation in infants according to delivery mode. Gut Microbes 2025; 17:2493900. [PMID: 40237336 PMCID: PMC12005435 DOI: 10.1080/19490976.2025.2493900] [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: 10/23/2024] [Revised: 01/29/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025] Open
Abstract
Exclusive breastfeeding (EBF) plays a crucial role in infant gut microbiome assembly and development. However, few studies have investigated the effects of EBF in restoring a perturbed microbiome. In this study, we applied whole metagenomic sequencing to assess the gut microbiome assembly in 525 Brazilian infants from 3 to 9 months of age of the Germina Cohort, demonstrating the early determinants of microbial taxonomy and function modulation. Our analysis shows that EBF alters the relative abundance of genes related to the microbiome taxonomy and function, with effects varying by delivery mode. EBF alters the pattern of carbohydrates, lipid metabolism, and cell structure pathways depending on the delivery mode. The microbiome age is closer to chronological infant age in EBF than in non-EBF infants, meaning a lower microbiome maturation index (MMI). Using a complementary machine learning approach, we show that Escherichia coli, Ruminococcus gnavus, and Clostridium neonatale, as well as vitamin K and o-antigen pathways contribute strongly to EBF prediction. Moreover, EBF influences the microbiome maturation in early life, toward a microbiome age more similar to the chronological infant's age.
Collapse
Affiliation(s)
| | - Paulo A. Schüroff
- School of Arts, Sciences and Humanity, University of Sao Paulo, Sao Paulo, Brazil
| | - Pedro A. R. Vanzele
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Davi Pereira-Santos
- Department of Applied Mathematics and Statistics, Institute of Mathematics and Computer Sciences, University of Sao Paulo, Sao Carlos, Brazil
- Departamento Acadêmico de Computação, Universidade Tecnológica Federal do Paraná (UTFPR), Câmpus Medianeira, Medianeira, Brazil
| | - Tamires Amabili Valim
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Kevin S. Bonham
- Department of Biological Sciences, Wellesley College, Wellesley, MA, USA
| | - André Fujita
- Division of Network AI Statistics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
- Department of Computer Science, Institute of Mathematics and Statistics, University of Sao Paulo, Sao Paulo, Brazil
| | - Maria Rita Passos-Bueno
- Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Patricia C. B. Beltrão-Braga
- Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
- Laboratory of Disease Modeling, Institut Pasteur de Sao Paulo, Sao Paulo, Brazil
| | - André C. P. L. F. Carvalho
- Department of Applied Mathematics and Statistics, Institute of Mathematics and Computer Sciences, University of Sao Paulo, Sao Carlos, Brazil
| | - Vanja Klepac-Ceraj
- Department of Biological Sciences, Wellesley College, Wellesley, MA, USA
| | - Guilherme V. Polanczyk
- Department of Psychiatry, Faculdade de Medicina FMUSP, University of Sao Paulo, Sao Paulo, Brazil
| | - Alline C. Campos
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Carla R. Taddei
- Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
- Division of Clinical Laboratory, University Hospital - University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
2
|
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
|
3
|
Bénet T, Dardinier A, Tytgat HLP, Austin S. Quantitative determination of human milk oligosaccharides in faecal matter. Anal Biochem 2025; 702:115845. [PMID: 40097060 DOI: 10.1016/j.ab.2025.115845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 03/05/2025] [Accepted: 03/12/2025] [Indexed: 03/19/2025]
Abstract
Human milk oligosaccharides (HMOs) are a major component of human milk and colostrum, yet they are non-digestible and thus not utilized directly by the infant. Nevertheless, they are important for infant health and development and have been implicated in immune development, pathogen deflection, cognitive development and the development of healthy microbiome. To understand how HMOs may be utilized it is important to be able to measure them both in milk and faeces. Many methods for the determination of HMOs in milk have been published. However, there are fewer reports of methods describing the quantitative determination of oligosaccharides in faeces. Here we report a validated method for the determination of 30 oligosaccharides in faeces. Oligosaccharides are labelled with 2-aminobenzamide and determined by liquid chromatography with fluorescence detection. The method precision determined as relative standard deviation under intermediate reproducibility conditions is below 12 % for all of the oligosaccharides. Recoveries were in the range 86.6-115 % for the 8 oligosaccharides for which quantitative standards were available, and are estimated to be in the range 81-117 % when using 2'-fucosyllactose as a universal calibrant assuming equimolar response factors of the 2-aminobenzamide labelled oligosaccharides.
Collapse
Affiliation(s)
- Thierry Bénet
- Nestlé Institute of Food Safety and Analytical Sciences, Nestlé Research, Lausanne, Switzerland
| | - Adrien Dardinier
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
| | - Hanne L P Tytgat
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
| | - Sean Austin
- Nestlé Institute of Food Safety and Analytical Sciences, Nestlé Research, Lausanne, Switzerland.
| |
Collapse
|
4
|
Hamouda HI, Li T, Shabana S, Hashem AH, Yin H. Advances in fucoidan and fucoidan oligosaccharides: Current status, future prospects, and biological applications. Carbohydr Polym 2025; 358:123559. [PMID: 40383599 DOI: 10.1016/j.carbpol.2025.123559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/10/2025] [Accepted: 03/27/2025] [Indexed: 05/20/2025]
Abstract
Sulfated polysaccharides (SPS) derived from seaweeds are precious bioactive compounds of diverse biological activities. Fucoidan is a complex SPS composed of L-fucose and sulfate groups, can be extracted from brown seaweeds, as well as microbial, insect, plant glycans, and marine invertebrates. It has gained considerable attention due to its anti-inflammatory, anticancer, antiviral, antithrombotic, hypolipidemic, and immune-modulatory properties. Recent research has focused on the extraction and extensive characterization of fucoidan. Its structural complexity, influenced by species, sources, and harvesting conditions, directly influences its bioactivity, with higher sulfation and lower molecular weight enhancing its activity. Interestingly, fucoidan oligosaccharides (FOs) play a critical role in various metabolic processes and hold significant potential in disease diagnostics. This comprehensive review explores the current status of fucoidan research, covering its sources, extraction and purification techniques, structural variations and biological activities. Additionally, we highlight its potential health benefits, providing insights for researchers interested in sulfated polysaccharides.
Collapse
Affiliation(s)
- Hamed I Hamouda
- Dalian Engineering Research Center for Carbohydrate Agricultural Preparations, Dalian Technology Innovation Center for Green Agriculture, Liaoning Provincial Key Laboratory of Carbohydrates, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; Processes Development Department, Egyptian Petroleum Research Institute (EPRI), Nasr City 11727, Cairo, Egypt
| | - Tang Li
- Dalian Engineering Research Center for Carbohydrate Agricultural Preparations, Dalian Technology Innovation Center for Green Agriculture, Liaoning Provincial Key Laboratory of Carbohydrates, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Samah Shabana
- School of Biomedical Engineering, Faculty of Medicine, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian 116024, China
| | - Amr H Hashem
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Cairo 11884, Egypt
| | - Heng Yin
- Dalian Engineering Research Center for Carbohydrate Agricultural Preparations, Dalian Technology Innovation Center for Green Agriculture, Liaoning Provincial Key Laboratory of Carbohydrates, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| |
Collapse
|
5
|
Al-Beltagi M. Human milk oligosaccharide secretion dynamics during breastfeeding and its antimicrobial role: A systematic review. World J Clin Pediatr 2025; 14. [DOI: 10.5409/wjcp.v14.i2.104797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 02/19/2025] [Accepted: 02/27/2025] [Indexed: 03/18/2025] Open
Abstract
BACKGROUND
Human milk oligosaccharides (HMOs) are bioactive components of breast milk with diverse health benefits, including shaping the gut microbiota, modulating the immune system, and protecting against infections. HMOs exhibit dynamic secretion patterns during lactation, influenced by maternal genetics and environmental factors. Their direct and indirect antimicrobial properties have garnered significant research interest. However, a comprehensive understanding of the secretion dynamics of HMOs and their correlation with antimicrobial efficacy remains underexplored.
AIM
To synthesize current evidence on the secretion dynamics of HMOs during lactation and evaluate their antimicrobial roles against bacterial, viral, and protozoal pathogens.
METHODS
A systematic search of PubMed, Scopus, Web of Science, and Cochrane Library focused on studies investigating natural and synthetic HMOs, their secretion dynamics, and antimicrobial properties. Studies involving human, animal, and in vitro models were included. Data on HMO composition, temporal secretion patterns, and mechanisms of antimicrobial action were extracted. Quality assessment was performed using validated tools appropriate for study design.
RESULTS
A total of 44 studies were included, encompassing human, animal, and in vitro research. HMOs exhibited dynamic secretion patterns, with 2′-fucosyllactose (2′-FL) and lacto-N-tetraose peaking in early lactation and declining over time, while 3-fucosyllactose (3-FL) increased during later stages. HMOs demonstrated significant antimicrobial properties through pathogen adhesion inhibition, biofilm disruption, and enzymatic activity impairment. Synthetic HMOs, including bioengineered 2′-FL and 3-FL, were structurally and functionally comparable to natural HMOs, effectively inhibiting pathogens such as Pseudomonas aeruginosa , Escherichia coli , and Campylobacter jejuni . Additionally, HMOs exhibited synergistic effects with antibiotics, enhancing their efficacy against resistant pathogens.
CONCLUSION
HMOs are vital in antimicrobial defense, supporting infant health by targeting various pathogens. Both natural and synthetic HMOs hold significant potential for therapeutic applications, particularly in infant nutrition and as adjuncts to antibiotics. Further research, including clinical trials, is essential to address gaps in knowledge, validate findings, and explore the broader applicability of HMOs in improving maternal and neonatal health.
Collapse
Affiliation(s)
- Mohammed Al-Beltagi
- Department of Paediatrics, Faculty of Medicine, Tanta University, Tanta 31511, Alghrabia, Egypt
- Department of Pediatric, University Medical Center, King Abdulla Medical City, Arabian Gulf University, Manama 26671, Bahrain
| |
Collapse
|
6
|
Liu P, Chen X, Cao X, Wang Y, Gao Y, Xu L, Jiang X, Xiao M. Semi-rational engineering of an α-L-fucosidase for regioselective synthesis of fucosyl- N-acetylglucosamine disaccharides. FOOD CHEMISTRY. MOLECULAR SCIENCES 2025; 10:100244. [PMID: 40034538 PMCID: PMC11875152 DOI: 10.1016/j.fochms.2025.100244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 01/30/2025] [Accepted: 02/07/2025] [Indexed: 03/05/2025]
Abstract
α-L-Fucosidases are attractive biocatalysts for the production of bioactive fucosylated oligosaccharides, however, poor regioselectivity and activity for transglycosylation have significantly limited their applications. We have recently derived an α-L-Fucosidase, BF3242, from Bacteroides fragilis NCTC9343, which could efficiently synthesize a mixture of Fuc-α-1,3/1,6-GlcNAc, but its 1,3/1,6-regioselectivity was observably affected by reaction temperature. Here, we integrated loop-targeted random mutagenesis and site-directed mutagenesis to engineer the regioselectivity and transglycosylation activity of BF3242. Loop-targeted random mutagenesis revealed that L266 in the loop-4 (H242-S267) within the model of BF3242 was a key residue for the regioselectivity for transglycosylation, and the saturation mutagenesis at residue L266 uncovered a mutant L266H with a significantly increased 1,3-regioselectivity of 97 % from 69 % of WT BF3242. Subsequently, five designed single-site mutations at the putative aglycone subsites were performed, resulting in a double-site mutant L266H/M285C that increased the overall yield of Fuc-α-1,3/1,6-GlcNAc to 76 % from 68 % of WT BF3242. The saturation mutagenesis at residue M285 finally generated a double-site mutant L266H/M285T with the maximal overall yield of Fuc-α-1,3/1,6-GlcNAc of 85 % and 1,3-regioselectivity of 98 %. The R T/H of L266H/M285T was approximately 2.7-fold higher than that of the WT BF3242. Molecular dynamics simulations revealed that the structural flexibility of the loop-4 was substantially reduced in mutant L266H, and the hydrogen bond formation and binding affinity between mutant L266H/M285T and Fuc-α-1,3-GlcNAc was significantly enhanced. The semi-rationally engineered enzyme L266H/M285T would be a promising biocatalyst for highly 1,3-regioselective synthesis of fucosyl-N-acetylglucosamine disaccharide.
Collapse
Affiliation(s)
- Peng Liu
- State Key Laboratory of Microbial Technology, National Glycoengineering Research Center, Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao 266237, China
- Department of Clinical Laboratory, Jinan Maternity and Child Care Hospital Affiliated to Shandong First Medical University, Jinan 250000, China
| | - Xiaodi Chen
- State Key Laboratory of Microbial Technology, National Glycoengineering Research Center, Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao 266237, China
- Department of Clinical Laboratory, Jinan Maternity and Child Care Hospital Affiliated to Shandong First Medical University, Jinan 250000, China
| | - Xueting Cao
- State Key Laboratory of Microbial Technology, National Glycoengineering Research Center, Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao 266237, China
| | - Yuying Wang
- State Key Laboratory of Microbial Technology, National Glycoengineering Research Center, Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao 266237, China
| | - Yafei Gao
- State Key Laboratory of Microbial Technology, National Glycoengineering Research Center, Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao 266237, China
| | - Li Xu
- State Key Laboratory of Microbial Technology, National Glycoengineering Research Center, Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao 266237, China
| | - Xukai Jiang
- State Key Laboratory of Microbial Technology, National Glycoengineering Research Center, Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao 266237, China
| | - Min Xiao
- State Key Laboratory of Microbial Technology, National Glycoengineering Research Center, Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao 266237, China
| |
Collapse
|
7
|
Becerra JE, Gallego Del Sol F, Rubio-Del-Campo A, Rodríguez-Díaz J, Monedero V, Marina A, Yebra MJ. Unveiling the structural bases of α-L-fucosidase B activity through mutants boosting transfucosylation efficiency. Int J Biol Macromol 2025; 311:143462. [PMID: 40286956 DOI: 10.1016/j.ijbiomac.2025.143462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 04/11/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
The AlfB α-L-fucosidase from Lacticaseibacillus paracasei exhibits high specificity on fucosyl-α1,3-N-acetylglucosamine, achieving yields of 30 % in transfucosylation reactions for its synthesis. By random mutagenesis we selected AlfB variants with enhanced transfucosylation activity. Expression of a collection of alfB mutants in E. coli resulted in the isolation of eighteen clones with reduced activity on p-nitrophenyl-α-L-fucopyranoside. The AlfB variants carried diverse amino substitutions, leading to modifications in their enzymatic parameters. In some cases, these changes increased transfucosylation yields, although no direct correlation was observed between kcat or Km values and the yields. One particular AlfB mutant (M58) achieved 100 % yield in the synthesis of fucosyl-α1,3-N-acetylglucosamine. This enzyme contained three amino acid substitutions (N196S, V261M and N346K); however, further analysis confirmed that the N346K mutation was sufficient to generate the maximum yield. Elucidation of the tridimensional structure of AlfB and AlfBM58 through X-ray crystallography allowed us to propose a mechanism by which the mutation at position 346, located in a loop close to the active site of an adjacent monomer in the protein tetramer, enhanced transfucosylation over hydrolysis of fucosyl-α1,3-N-acetylglucosamine. This study paves the way for designing novel AlfB variants as tools for the efficient enzymatic synthesis of regio-specific fucosyl-oligosaccharides of biotechnological interest.
Collapse
Affiliation(s)
- Jimmy E Becerra
- Laboratorio de Bacterias Lácticas y Probióticos, Departamento de Biotecnología de Alimentos, Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Valencia, Spain
| | - Francisca Gallego Del Sol
- Departamento de Genómica y Proteómica, Instituto de Biomedicina de Valencia (IBV-CSIC), Valencia, Spain.
| | - Antonio Rubio-Del-Campo
- Laboratorio de Bacterias Lácticas y Probióticos, Departamento de Biotecnología de Alimentos, Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Valencia, Spain
| | - Jesús Rodríguez-Díaz
- Departamento de Microbiología, Facultad de Medicina, Universitat de València, Valencia, Spain
| | - Vicente Monedero
- Laboratorio de Bacterias Lácticas y Probióticos, Departamento de Biotecnología de Alimentos, Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Valencia, Spain.
| | - Alberto Marina
- Departamento de Genómica y Proteómica, Instituto de Biomedicina de Valencia (IBV-CSIC), Valencia, Spain
| | - María J Yebra
- Laboratorio de Bacterias Lácticas y Probióticos, Departamento de Biotecnología de Alimentos, Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Valencia, Spain
| |
Collapse
|
8
|
Hunt KE, Miller A, Jarg T, Kriis K, Kanger T. Selective Acetylation of Unprotected Thioglycosides and Fully Unprotected Monosaccharides with Candida antarctica Lipase‑B. ACS OMEGA 2025; 10:20047-20053. [PMID: 40415852 PMCID: PMC12096258 DOI: 10.1021/acsomega.5c02467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/14/2025] [Accepted: 04/29/2025] [Indexed: 05/27/2025]
Abstract
A selective enzymatic acetylation method for the protection of the second and the sixth positions of thio-d-galactopyranoside was found using immobilized Candida antarctica lipase-B (CAL-B). Unfortunately, it was determined that the immobilized enzyme cannot be recycled effectively. The optimized acetylation method was screened with different thioglycosides and with fully unprotected saccharides. New methods for several new partially protected saccharides were found, while the synthesis of some known saccharides, e.g., the third and the sixth position-protected d-glucose or the fourth position-protected l-rhamnose, was improved. Furthermore, an enzymatic acetal formation between the fourth and the sixth positions was discovered. The main limitation for acetylation reactions with CAL-B has been determined to be the substrate solubility.
Collapse
Affiliation(s)
- Kaarel Erik Hunt
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, Tallinn12618, Estonia
| | - Annette Miller
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, Tallinn12618, Estonia
| | - Tatsiana Jarg
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, Tallinn12618, Estonia
| | - Kadri Kriis
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, Tallinn12618, Estonia
| | - Tõnis Kanger
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, Tallinn12618, Estonia
| |
Collapse
|
9
|
Tang J, Shen C, Yao D, Yu J, Liu Y, Tu M, Zhang H, Xu X, Lai OM, Cheong LZ. Differences in mature human milk metabolic profiles based on delivery mode and parity. J Nutr Biochem 2025:109967. [PMID: 40398823 DOI: 10.1016/j.jnutbio.2025.109967] [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: 07/08/2024] [Revised: 03/20/2025] [Accepted: 05/16/2025] [Indexed: 05/23/2025]
Abstract
Human milk (HM) is regarded as the gold standard for infant nutrition. The metabolite profiles of mature human milk (MHM) have been reported to be affected by maternal physiological conditions, lactation stage, and maternal diets. We collected MHM (3-6 months postpartum) from 32 healthy mothers with different parities and delivery modes. Then, GC-MS and LC-MS were used to perform an untargeted metabolomics study. A clear distinction can be observed in terms of MHM metabolites of mothers with different delivery modes and parities with a 95% confidence interval. A total of 119 differentially expressed metabolites (DEMs) were identified in MHM of women with different delivery modes. Metabolic pathway analyses indicated that these DEMs are mainly associated with fatty acid biosynthesis. The higher abundances of these DEMs in MHM of cesarean women may be due to the differing levels of cortisol and oxytocin between mothers with different delivery modes. Meanwhile, 284 DEMs were identified in MHM of women with different parities. These DEMs are primarily related to ABC transporters, center carbon metabolism in cancer, and D-amino acid metabolism. These findings highlighted the impact of delivery modes and parity on HM metabolite composition. Further research is needed to explore the long-term health implications of these metabolic differences and optimize infant nutrition strategies.
Collapse
Affiliation(s)
- Jiayue Tang
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, College of Food Science and Engineering, Ningbo University, Ningbo, 315211, China
| | - Cai Shen
- School of Agriculture, Food and Ecosystem Sciences, Faculty of Science, University of Melbourne, 3010, Australia
| | - Dan Yao
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, College of Food Science and Engineering, Ningbo University, Ningbo, 315211, China
| | - Jingwen Yu
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, College of Food Science and Engineering, Ningbo University, Ningbo, 315211, China
| | - Yanan Liu
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, College of Food Science and Engineering, Ningbo University, Ningbo, 315211, China
| | - Maolin Tu
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, College of Food Science and Engineering, Ningbo University, Ningbo, 315211, China
| | - Hong Zhang
- Wilmar (Shanghai) Biotechnology Research and Development Center Co Ltd., No.118 Gaodong Rd., Pudong New District, Shanghai 200137, China
| | - Xuebing Xu
- Wilmar (Shanghai) Biotechnology Research and Development Center Co Ltd., No.118 Gaodong Rd., Pudong New District, Shanghai 200137, China
| | - Oi-Ming Lai
- Department of Bioprocess Technology, Faculty of Biotechnology and Bimolecular Science, University Putra Malaysia UPM, 43400, Serdang, Selangor, Malaysia
| | - Ling-Zhi Cheong
- School of Agriculture, Food and Ecosystem Sciences, Faculty of Science, University of Melbourne, 3010, Australia.
| |
Collapse
|
10
|
Hellinga AH, Zuurveld M, Mank M, Kraneveld AD, Garssen J, Spann K, Bode L, Willemsen LEM, van’t Land B. Secretor and non-secretor human milk oligosaccharides differentially modulate immune response in the presence of cow's milk allergen β-lactoglobulin in an in vitro sensitization model. Front Immunol 2025; 16:1575656. [PMID: 40416953 PMCID: PMC12098339 DOI: 10.3389/fimmu.2025.1575656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/17/2025] [Indexed: 05/27/2025] Open
Abstract
Introduction Food allergies, like cow's milk allergy, significantly impact children, with sensitization often beginning during the first year of life. Human milk oligosaccharides (HMOs) may influence this process, as specific HMOs differentially affect mucosal immune responses in vitro. Given the distinct HMO profiles of secretor (Se+) and non-secretor (Se-) milk, we investigate how the full HMO profiles from Se+ and Se- milk affect immune responses in the absence or presence of a cow's milk allergen. Methods Monocyte-derived dendritic cells (moDCs) were exposed to isolated Se+ and Se- pooled HMOs (pHMOs), and subsequently co-cultured with naïve T cells to confirm immune modulation. We compared the type 2-activation capability of several cow's milk proteins via direct exposure to moDCs or via intestinal epithelial cells (IECs) co-cultured with moDCs. Finally, we studied the effect of pHMOs in the presence of cow's milk allergen β-lactoglobulin (BLG) (via (IECs)) on moDCs and subsequent T cell response. Results Both Se+ and Se- pHMOs dose-dependently activated moDCs, indicated by increased IL8 release and %CD80+ moDCs. Se+ pHMOs tended to increase type 2-associated markers, while also increasing regulatory IL10 release. Se+ pHMOs-pre-exposed moDCs instructed T cells to produce type 2 cytokines like IL13. Se- pHMOs reduced the %CD86+ moDCs but did not drive a type 2 signature in T cells. In the presence of BLG, Se+ pHMOs-pre-exposed moDCs also instructed IL13 release by T cells, while increasing the percentage regulatory T cells. In contrast, co-exposure of BLG with Se- pHMOs only slightly affected moDC phenotype, and these moDCs did not modify T cell phenotypes. Conclusions Se+ and Se- pHMOs with BLG differentially affected moDC activation. Se+ pHMO-pre-exposed moDCs induced a type 2- and regulatory-associated T cell phenotype. These data suggest that depending on the secretor status, HMOs differentially modulate immune responsiveness in vitro.
Collapse
Affiliation(s)
- Anneke H. Hellinga
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, Netherlands
| | - Marit Zuurveld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, Netherlands
| | - Marko Mank
- Global Center of Excellence Human Milk Research and Analytics, Danone Global Research and Innovation Center, Utrecht, Netherlands
| | - Aletta D. Kraneveld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, Netherlands
- Department of Neuroscience, Faculty of Science, Vrije Universiteit (VU) University, Amsterdam, Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, Netherlands
- Global Center of Excellence Immunology, Danone Global Research and Innovation Center, Utrecht, Netherlands
| | - Kennedy Spann
- Department of Pediatrics, Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (MOMI CORE), and the Human Milk Institute (HMI), University of California San Diego, La Jolla, CA, United States
| | - Lars Bode
- Department of Pediatrics, Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (MOMI CORE), and the Human Milk Institute (HMI), University of California San Diego, La Jolla, CA, United States
| | - Linette E. M. Willemsen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, Netherlands
| | - Belinda van’t Land
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
- Global Center of Excellence Immunology, Danone Global Research and Innovation Center, Utrecht, Netherlands
| |
Collapse
|
11
|
Brandt MJV, van Steenwinckel J, van Emst BL, Lohr J, Mank M, Schipper L, Harvey L, Benders MJNL, de Theije CGM. Human milk oligosaccharides improve white matter and interneuron development in a double-hit rat model for preterm brain injury. Neuropharmacology 2025; 276:110507. [PMID: 40350143 DOI: 10.1016/j.neuropharm.2025.110507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/27/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
Mother's own milk is the preferred source of nutrition for preterm infants due to its beneficial compounds, including human milk oligosaccharides (HMOs). HMOs support microbiota and immune development, but their effect on the preterm brain remains unstudied. Here, we examined the therapeutic potential of HMOs and short-chain galacto- and long-chain fructo-oligosaccharides (scGOS/lcFOS) in a preclinical model for encephalopathy of prematurity. Pregnant Wistar rats were injected with 10 μg/kg lipopolysaccharides at embryonic day 20, and pups were exposed to hypoxia (8% O2, 140 min) at postnatal day (P)4 (fetal inflammation and postnatal hypoxia; FIPH). From P1, FIPH-pups of both sexes were treated intragastrically with HMOs, scGOS/lcFOS (9:1), or water. Transcriptomic analysis of CD11b/c + microglia was performed at P6, while immunohistochemical, microbial and short-chain fatty acid (SCFA) analyses were performed at P20. Decreased cortical myelin in FIPH animals was rescued exclusively by HMOs. Furthermore, both HMOs and scGOS/lcFOS treatments normalized reduced parvalbumin+ interneuron numbers in the hippocampus, potentially through promoting beneficial bacteria, including Lactobacillus and Bifidobacterium, and cecal acetic acid content. Interestingly, treatment with HMOs more effectively restored FIPH-induced upregulation of microglial genes associated with immune activation and normalized persistent activated microglial morphology in FIPH-males. HMOs supplementation holds promise to improve neurodevelopmental outcomes following preterm birth.
Collapse
Affiliation(s)
- Myrna J V Brandt
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Lundlaan 6, 3584 EA, Utrecht, the Netherlands.
| | - Juliette van Steenwinckel
- Université Paris Cité, Inserm, NeuroDiderot, FHU Prem'impact, 48 Bd Sérurier, F-75019, Paris, France.
| | - Bobbie-Louise van Emst
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Lundlaan 6, 3584 EA, Utrecht, the Netherlands.
| | - Julia Lohr
- Danone Research and Innovation, Uppsalalaan 12, 3584 HD, Utrecht, the Netherlands.
| | - Marko Mank
- Danone Research and Innovation, Uppsalalaan 12, 3584 HD, Utrecht, the Netherlands.
| | - Lidewij Schipper
- Danone Research and Innovation, Uppsalalaan 12, 3584 HD, Utrecht, the Netherlands.
| | - Louise Harvey
- Danone Research and Innovation, Uppsalalaan 12, 3584 HD, Utrecht, the Netherlands.
| | - Manon J N L Benders
- Department of Neonatology, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Lundlaan 6, 3584 EA, Utrecht, the Netherlands
| | - Caroline G M de Theije
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Lundlaan 6, 3584 EA, Utrecht, the Netherlands.
| |
Collapse
|
12
|
Pang J, Sa Z, Zhao X, Li J, Bai G, Xia Y. Human Milk Oligosaccharide Lacto- N-Neotetraose Promotes Gut Microbiota Recovery in the Context of Antibiotic-Induced Dysbiosis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:10331-10346. [PMID: 40244944 DOI: 10.1021/acs.jafc.5c01911] [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: 04/19/2025]
Abstract
Human milk oligosaccharides (HMOs) may shape intestinal homeostasis, although the optimal form of HMOs to restore the gut microbiota in antibiotic-induced dysbiosis remains unclear. Here, we found that HMOs with various structures modulate microbial communities differently after antibiotic exposure. Lacto-N-neotetraose (LNnT) better promotes the recovery of intestinal microbiota (chiefly Lactobacillus) and increases the level of Bifidobacterium compared to 3'-sialyllactose, 2'-fucosyllactose, and the mixture. Additionally, LNnT decreases the potential pathogenic bacteria Klebsiella level and the microbial dysbiosis index. Although supplementation with LNnT does not decrease the Clostridioides difficile burden or alleviate the decline in the fecal numbers of Lactobacillus and Bifidobacterium after C. difficile infection (CDI), LNnT attenuates intestinal epithelial damage, decreases inflammatory status, and alters metabolome profiles after CDI. Collectively, LNnT may function as a promising prebiotic to promote gut microbiota recovery in the context of antibiotic-induced dysbiosis.
Collapse
Affiliation(s)
- Jiaman Pang
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Zhixuan Sa
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Xuan Zhao
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Jiawei Li
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Guangdong Bai
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Yaoyao Xia
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| |
Collapse
|
13
|
Huang Z, Luo S, Li Y, Li Z, Yi C, Zhang Y, Hu Y, Chen B. Impact of Maternal Metabolic Status on Human Milk Oligosaccharide Composition: A Population-Based Cross-Sectional Study in Central South China. Nutrients 2025; 17:1480. [PMID: 40362789 PMCID: PMC12073883 DOI: 10.3390/nu17091480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2025] [Revised: 04/23/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Background: Human milk oligosaccharides (HMOs) serve as critical bioactive components supporting infant growth and development. However, the influence of maternal metabolic factors during lactation on HMOs remains to be fully elucidated. This study aimed to investigate the association between maternal metabolic factors and HMOs, as well as the potential mediating effects of these factors. Methods: An observational cross-sectional study was conducted in Central South China, enrolling 196 lactating mothers. HMOs were quantified using liquid chromatography-tandem mass spectrometry. Maternal metabolic factors were assessed through physical examinations. Associations between metabolic factors and HMOs were analyzed using linear regression, and mediation effects were evaluated. Results: HMOs from Central South China were predominantly composed of neutral fucosylated HMOs. Significant differences were observed in the levels of several HMOs across maternal age groups and lactation periods. The concentration of 3'-sialyllactose (3'-SL) exhibited a negative association with the pre-pregnancy body mass index (BMI) (β = -0.16, 95% CI: -0.29, -0.03; p = 0.02), while a positive association was found with maternal heart rate (β = 0.14, 95% CI: 0.01, 0.27; p = 0.04). However, these associations were different between secretor and non-secretor mothers. Associations of 3'-SL with pre-pregnancy BMI and maternal HR were only found in the secretor mothers. Triglycerides and low-density lipoprotein cholesterol mediated the associations between maternal pre-pregnancy BMI and 3'-sialyllactose (3'-SL). Conclusions: The variations of several HMOs among mothers from Central South China were associated with maternal age and lactation period. The concentration of 3'-SL was negatively correlated with maternal pre-pregnancy BMI. The potential mechanism underlying the influence of maternal BMI on 3'-SL levels may involve maternal lipid metabolism and genetic factors.
Collapse
Affiliation(s)
- Zhi Huang
- School of Medical Laboratory, Hunan University of Medicine, Jinxi Road No. 492, Huaihua 418000, China; (Z.H.); (Y.Z.)
| | - Shurong Luo
- School of Chemistry & Chemical Engineering, Hunan Normal University, Lu Mountain Road No. 286, Changsha 410081, China;
| | - Yuxin Li
- Hunan Institute for Drug Control, Jinxing Middle Road No. 469, Yuelu District, Changsha 410001, China;
| | - Ziming Li
- The Department of Toxicology, Hunan Provincial Center for Disease Control and Prevention, Xinglian Road No. 861, Laodaohe Street, Kaifu District, Changsha 410005, China; (Z.L.); (C.Y.)
| | - Chuanzhu Yi
- The Department of Toxicology, Hunan Provincial Center for Disease Control and Prevention, Xinglian Road No. 861, Laodaohe Street, Kaifu District, Changsha 410005, China; (Z.L.); (C.Y.)
| | - Yan Zhang
- School of Medical Laboratory, Hunan University of Medicine, Jinxi Road No. 492, Huaihua 418000, China; (Z.H.); (Y.Z.)
| | - Yuming Hu
- The Department of Toxicology, Hunan Provincial Center for Disease Control and Prevention, Xinglian Road No. 861, Laodaohe Street, Kaifu District, Changsha 410005, China; (Z.L.); (C.Y.)
| | - Bo Chen
- School of Chemistry & Chemical Engineering, Hunan Normal University, Lu Mountain Road No. 286, Changsha 410081, China;
| |
Collapse
|
14
|
van Neerven RJJ. Macronutrients, Micronutrients, and Malnutrition: Effects of Nutrition on Immune Function in Infants and Young Children. Nutrients 2025; 17:1469. [PMID: 40362777 PMCID: PMC12073586 DOI: 10.3390/nu17091469] [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] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/16/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
The function of the immune system is not only dependent on factors like genetics, age, the environment, and exposure to infectious agents and allergens but also on our microbiota and our diet. It has been known for centuries that food can influence health and vulnerability to infection. This is especially true for infants, young children, and the elderly. This review focuses on how nutrition can support immune function from gestation to school-aged children. Immune support begins during pregnancy by the mother's diet and transfer of nutritional components as well as antibodies to her fetus. After birth, breastfeeding is of crucial importance for immune development as well as for the development of the intestinal microbiota of an infant. If breastfeeding is not possible, early-life formulas are an alternative. These can provide several of the functionalities of breastmilk, as well as the key nutrients a child needs. New foods are introduced during and after weaning, and after this period, children switch to consuming a normal diet. However, due to circumstances, children can be malnourished. This can range from severe protein/energy malnutrition to micronutrient deficiencies and obesity, all of which can affect the function of the immune system. This narrative review describes the immune challenges in early life, explores breastfeeding and early life nutrition, and provides mechanistic insight into the relative contribution of macronutrients, micronutrients and other immunomodulatory food components that can support immune function in early life.
Collapse
Affiliation(s)
- R J Joost van Neerven
- Cell Biology and Immunology, Wageningen University & Research, 6708 WD Wageningen, The Netherlands
| |
Collapse
|
15
|
Pooladian F, Das A, Demchenko AV. Chemical Synthesis of Two Fucosylated Human Milk Oligosaccharides: 3-Fucosyllactose and Lacto-N-fucopentaose V. Chemistry 2025:e202500754. [PMID: 40261993 DOI: 10.1002/chem.202500754] [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: 02/26/2025] [Revised: 04/18/2025] [Accepted: 04/22/2025] [Indexed: 04/24/2025]
Abstract
Glycans present in human milk have been the focus of many studies due to challenges associated with their synthesis. The development of new methods for obtaining individual glycans found in human milk has been a vibrant area of research in glycosciences. This study reports the synthesis of two fucosylated glycans found in human milk, 3-fucosyllactose, which was previously synthesized by both chemical and enzymatic methods, and the first chemical synthesis of lacto-N-fucopentaose V. Screening different protecting and leaving groups, as well as optimizing the glycosylation reaction conditions helped us to achieve efficient assembly and deprotection of these two fucosylated human milk oligosaccharides in good yields.
Collapse
Affiliation(s)
- Faranak Pooladian
- Department of Chemistry, Saint Louis University, 3501 Laclede Ave, St. Louis, Missouri, 63103, USA
| | - Anupama Das
- Department of Chemistry, Saint Louis University, 3501 Laclede Ave, St. Louis, Missouri, 63103, USA
| | - Alexei V Demchenko
- Department of Chemistry, Saint Louis University, 3501 Laclede Ave, St. Louis, Missouri, 63103, USA
| |
Collapse
|
16
|
Duan Q, Yu B, Huang Z, Luo Y, Zheng P, Mao X, Yu J, Luo J, Yan H, He J. Sialyllactose Attenuates Inflammation and Injury of Intestinal Epithelial Cells upon Enterotoxigenic Escherichia coli Infection. Int J Mol Sci 2025; 26:3860. [PMID: 40332525 PMCID: PMC12027521 DOI: 10.3390/ijms26083860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/11/2025] [Accepted: 04/15/2025] [Indexed: 05/08/2025] Open
Abstract
Sialyllactose (SL), a bioactive trisaccharide abundant in porcine colostrum, demonstrates multifunctional properties including antimicrobial activity, immune regulation, and apoptosis inhibition. This research uncovers the mechanisms by which SL mitigates enterotoxigenic Escherichia coli (ETEC)-mediated damage to intestinal barrier integrity, employing IPEC-J2 porcine epithelial models. SL pre-treatment effectively blocked pathogen adhesion by competitively binding to cellular receptors, concurrently mitigating inflammation through significant suppression of TNF-α, IL-1β, and IL-6 expression (p < 0.05). Notably, SL exhibited functional parallels to the NF-κB inhibitor BAY11-7082, jointly enhancing tight junction integrity via ZO-1 protein stabilization and inhibiting pro-inflammatory signaling through coordinated suppression of IκB-α/NF-κB phosphorylation cascades. The dual-action mechanism combines molecular interception of microbial attachment with intracellular modulation of the TLR4/MyD88/NF-κB pathway, effectively resolving both pathogenic colonization and inflammatory amplification. These findings position SL as a potential therapeutic application nutraceutical for livestock, with the capacity to address post-weaning porcine enteritis through functional feed formulations that synergistically enhance intestinal barrier resilience while curbing ETEC-mediated inflammatory pathogenesis.
Collapse
Affiliation(s)
- Qiming Duan
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.D.)
- Key Laboratory for Animal Disease-Resistance Nutrition, Chengdu 611130, China
| | - Bing Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.D.)
- Key Laboratory for Animal Disease-Resistance Nutrition, Chengdu 611130, China
| | - Zhiqing Huang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.D.)
- Key Laboratory for Animal Disease-Resistance Nutrition, Chengdu 611130, China
| | - Yuheng Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.D.)
- Key Laboratory for Animal Disease-Resistance Nutrition, Chengdu 611130, China
| | - Ping Zheng
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.D.)
- Key Laboratory for Animal Disease-Resistance Nutrition, Chengdu 611130, China
| | - Xiangbing Mao
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.D.)
- Key Laboratory for Animal Disease-Resistance Nutrition, Chengdu 611130, China
| | - Jie Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.D.)
- Key Laboratory for Animal Disease-Resistance Nutrition, Chengdu 611130, China
| | - Junqiu Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.D.)
- Key Laboratory for Animal Disease-Resistance Nutrition, Chengdu 611130, China
| | - Hui Yan
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.D.)
- Key Laboratory for Animal Disease-Resistance Nutrition, Chengdu 611130, China
| | - Jun He
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.D.)
- Key Laboratory for Animal Disease-Resistance Nutrition, Chengdu 611130, China
| |
Collapse
|
17
|
Wang Z, Zhang Z, Li Y, Mao Y, Wan Z, Zhang H. Two-step Production Method for Lacto- N-Triose II via Cell-Coupled Biocatalytic Strategy. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:8391-8400. [PMID: 40131335 DOI: 10.1021/acs.jafc.4c12052] [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: 03/26/2025]
Abstract
Lacto-N-triose II (LNT II) represents a significant neutral human milk oligosaccharide (HMO) that functions as a fundamental structural framework for the synthesis of more complex HMOs. In this study, we utilize GlcNAc and lactose as substrates, employing a whole-cell catalytic approach for the synthesis of LNT II. First, we have identified and successfully expressed three genes associated with the production of LNT II: Nahk, EcGlmU, and LgtA, synthesized LNT II through a whole-cell catalytic system utilizing multistrain coupling. Then, to further enhance the yield, we incorporated yeast cells for energy regeneration, employed coexpression strategies to minimize cell density, following a series of systematic optimizations resulting in a 7-fold increase in LNT II production. Finally, a two-step catalytic process was conducted in a 5L bioreactor, and the maximum LNT II production reached 52.34 g/L with a conversion rate of lactose 95.95%.
Collapse
Affiliation(s)
- Zhijie Wang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Zimeng Zhang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Yu Li
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Yijie Mao
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Zecheng Wan
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Hongtao Zhang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| |
Collapse
|
18
|
Zhang Y, Han T, Zhang L, Yun S, Yuan Y, Wang X, Huang L, Wang Z, Lu Y. Unveiling the N-Glycomic Diversity of Goat Lactoferrin during Lactation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:8515-8530. [PMID: 40153570 DOI: 10.1021/acs.jafc.4c10346] [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: 03/30/2025]
Abstract
Goat lactoferrin is an N-glycoprotein, and its glycan moieties are essential for its biological activity. However, the structures of these glycans, particularly the sialylated isomers with α-2,3- or α-2,6-linkages, remain poorly characterized. Utilizing online hydrophilic liquid chromatography-tandem mass spectrometry, our study characterized N-glycans in goat lactoferrin across different lactation stages, and the putative structures of the 86 N-glycans of goat lactoferrin were presented, including 53 previously undetected ones. The content and variety of N-glycans decreased from colostrum to mature milk, with transitional milk exhibiting the highest levels of neutral and high-mannose N-glycans. Colostrum was particularly rich in fucose- and sialylated N-glycans, especially those with α-2,6-linkages. Notably, the α-2,6-linked sialylated N-glycan H5N4F1A1-1 was 7.09-fold and 12.85-fold more abundant in colostrum compared to transitional and mature milk, respectively. These findings provide valuable insights into the structure of lactoferrin and could facilitate the development of functional goat milk products.
Collapse
Affiliation(s)
- Yuyang Zhang
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, Key Laboratory of Glycobiology and Glycoengineering of Xi'an, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Tianjiao Han
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, Key Laboratory of Glycobiology and Glycoengineering of Xi'an, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Lan Zhang
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, Key Laboratory of Glycobiology and Glycoengineering of Xi'an, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Shuai Yun
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, Key Laboratory of Glycobiology and Glycoengineering of Xi'an, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Yue Yuan
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, Key Laboratory of Glycobiology and Glycoengineering of Xi'an, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Xiaoqin Wang
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, Key Laboratory of Glycobiology and Glycoengineering of Xi'an, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Linjuan Huang
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, Key Laboratory of Glycobiology and Glycoengineering of Xi'an, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Zhongfu Wang
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, Key Laboratory of Glycobiology and Glycoengineering of Xi'an, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Yu Lu
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, Key Laboratory of Glycobiology and Glycoengineering of Xi'an, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| |
Collapse
|
19
|
Zhou B, Garber JM, Butcher J, Muszynski A, Casey RL, Huynh S, Archer-hartmann S, Porfírio S, Rogers AM, Azadi P, Parker CT, Ng KKS, Hines KM, Stintzi A, Szymanski CM. Campylobacter jejuni resistance to human milk involves the acyl carrier protein AcpP. mBio 2025; 16:e0399724. [PMID: 39998218 PMCID: PMC11980577 DOI: 10.1128/mbio.03997-24] [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: 01/13/2025] [Accepted: 01/28/2025] [Indexed: 02/26/2025] Open
Abstract
Campylobacter jejuni is a common foodborne pathogen worldwide that is associated with high rates of morbidity and mortality among infants in low- to middle-income countries (LMICs). Human milk provides infants with an important source of nutrients and contains antimicrobial components for protection against infection. However, recent studies, including our own, have found significantly higher levels of Campylobacter in diarrheal stool samples collected from breastfed infants compared to non-breastfed infants in LMICs. We hypothesized that C. jejuni has unique strategies to resist the antimicrobial properties of human milk. Transcriptional profiling found human milk exposure induces genes associated with ribosomal function, iron acquisition, and amino acid utilization in C. jejuni strains 81-176 and 11168. However, unidentified proteinaceous components of human milk prevent bacterial growth. Evolving both C. jejuni isolates to survive in human milk resulted in mutations in genes encoding the acyl carrier protein (AcpP) and the major outer membrane porin (PorA). Introduction of the PorA/AcpP amino acid changes into the parental backgrounds followed by electron microscopy showed distinct membrane architectures, and the AcpP changes not only significantly improved growth in human milk, but also yielded cells surrounded with outer membrane vesicles. Analyses of the phospholipid and lipooligosaccharide (LOS) compositions suggest an imbalance in acyl chain distributions. For strain 11168, these changes protect both evolved and 11168∆acpPG33R strains from bacteriophage infection and polymyxin killing. Taken together, this study provides insights into how C. jejuni may evolve to resist the bactericidal activity of human milk and flourish in the hostile environment of the gastrointestinal tract. IMPORTANCE In this study, we evolved C. jejuni strains which can grow in the presence of human milk and found that cell membrane alterations may be involved in resistance to the antimicrobial properties of human milk. These bacterial membrane changes are predominantly linked to amino acid substitutions within the acyl carrier protein, AcpP, although other bacterial components, including PorA, are likely involved. This study provides some insights into possible strategies for C. jejuni survival and propagation in the gastrointestinal tract of breastfed infants.
Collapse
Affiliation(s)
- Bibi Zhou
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Jolene M. Garber
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - James Butcher
- School of Pharmaceutical Sciences, Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Artur Muszynski
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Rebekah L. Casey
- Department of Chemistry, University of Georgia, Athens, Georgia, USA
| | - Steven Huynh
- Produce Safety and Microbiology Research Unit, Agricultural Research Service, U.S. Department of Agriculture, Albany, California, USA
| | | | - Sara Porfírio
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Ashley M. Rogers
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Craig T. Parker
- Produce Safety and Microbiology Research Unit, Agricultural Research Service, U.S. Department of Agriculture, Albany, California, USA
| | - Kenneth K. S. Ng
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario, Canada
| | - Kelly M. Hines
- Department of Chemistry, University of Georgia, Athens, Georgia, USA
| | - Alain Stintzi
- School of Pharmaceutical Sciences, Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Christine M. Szymanski
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
20
|
Hülsmeier AJ. Glycosphingolipids in neurodegeneration - Molecular mechanisms, cellular roles, and therapeutic perspectives. Neurobiol Dis 2025; 207:106851. [PMID: 39978484 DOI: 10.1016/j.nbd.2025.106851] [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: 12/19/2024] [Revised: 02/15/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025] Open
Abstract
Neurodegenerative diseases, including Alzheimer's (AD), Parkinson's (PD), Huntington's (HD), and amyotrophic lateral sclerosis (ALS), are characterized by progressive neuronal loss and pose significant global health challenges. Glycosphingolipids (GSLs), critical components of neuronal membranes, regulate signal transduction, membrane organization, neuroinflammation, and lipid raft functionality. This review explores GSL roles in neural development, differentiation, and neurogenesis, along with their dysregulation in neurodegenerative diseases. Aberrations in GSL metabolism drive key pathological features such as protein aggregation, neuroinflammation, and impaired signaling. Specific GSLs, such as GM1, GD3, and GM3, influence amyloid-beta aggregation in AD, α-synuclein stability in PD, and mutant huntingtin toxicity in HD. Therapeutic strategies targeting GSL metabolism, such as GM1 supplementation and enzyme modulation, have demonstrated potential to mitigate disease progression. Further studies using advanced lipidomics and glycomics may support biomarker identification and therapeutic advancements. This work aims to highlight the translational potential of GSL research for diagnosing and managing devastating neurodegenerative conditions.
Collapse
Affiliation(s)
- Andreas J Hülsmeier
- Institute of Clinical Chemistry, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
21
|
Park J, Jinno C, Wickramasinghe S, Mills DA, Liu Y, Lönnerdal BL, Ji P. Iron Fortification and Inulin Supplementation in Early Infancy: Evaluating the Impact on Gut Microbiome in a Piglet Model. Curr Dev Nutr 2025; 9:104587. [PMID: 40242393 PMCID: PMC12002768 DOI: 10.1016/j.cdnut.2025.104587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 03/03/2025] [Accepted: 03/08/2025] [Indexed: 04/18/2025] Open
Abstract
Background Prophylactic iron fortification in infant formula effectively prevents iron deficiency anemia. However, the low absorption rate results in excess unabsorbed iron accumulates in colon, where it has been linked to harmful microbiota changes and increased diarrheal incidence. Prebiotic oligosaccharides have shown promise in mitigating these adverse effects, but the role of inulin or synbiotic supplementation with inulin-fermenting lactic acid bacteria in modulating early gut microbiome under iron fortification remains understudied. Objectives This study used a neonatal pig model to investigate the effects of iron fortification and inulin supplementation, with or without Ligilactobacillus agilis YZ050 (L. agilis), on gut microbiome. Methods Twenty-four piglets were stratified and randomly assigned into 1 of the 4 dietary treatments from postnatal day (PD) 2: iron-adequate milk (AI), high-iron milk (HI), high-iron milk with 5% inulin (HIP), or HIP milk with oral gavage of L . agilis every third day (HIS). Piglets were individually housed and fed milk in proportion to body weight in 14 meals daily, simulating formula feeding in infants. Fecal and colonic microbiome were analyzed via 16S rRNA sequencing, with microbial diversity and relative abundance analyzed using QIIME2 and R. Results Iron fortification, regardless of inulin supplementation, decreased α-diversity compared with AI. β-Diversity showed clustering of HIP and HIS samples, which were distinct from AI and HI. Although iron fortification had minor impact on microbial composition, inulin supplementation significantly modified microbiome diversity, increasing Prevotella, Megasphaera, and Lachnospiraceae_NK3A20_group species, while reducing Bacteroides and Ruminococcus. Colonic microbiome shifted from Bacteroides-dominant enterotype in AI and HI groups to Prevotella-dominant enterotype in HIP and HIS groups, indicating enhanced fiber degrading capacity. Despite its inulin-fermenting property, L . agilis showed limited colonization and minimal microbiome impact. Conclusions Inulin supplementation significantly influenced gut microbiome, shifting enterotype from Bacteroides to Prevotella. dominance and overriding the effect of high-iron fortification in a milk-fed piglet model.
Collapse
Affiliation(s)
- Jungjae Park
- Department of Nutrition, University of California, Davis, CA, United States
| | - Cynthia Jinno
- Department of Animal Science, University of California, Davis, CA, United States
| | - Saumya Wickramasinghe
- Department of Food Science and Technology, University of California, Davis, CA, United States
| | - David A Mills
- Department of Food Science and Technology, University of California, Davis, CA, United States
| | - Yanhong Liu
- Department of Animal Science, University of California, Davis, CA, United States
| | - Bo L Lönnerdal
- Department of Nutrition, University of California, Davis, CA, United States
| | - Peng Ji
- Department of Nutrition, University of California, Davis, CA, United States
| |
Collapse
|
22
|
Tunç C, Dogan AK. The Effects of "Oral Administration of Breast Milk Droplets" and "Palatal Stimulation with a Finger" Methods on Feed Tolerance in Preterm Newborns Fed via an Orogastric Tube: Randomized Controlled Trial. Niger J Clin Pract 2025; 28:452-460. [PMID: 40289001 DOI: 10.4103/njcp.njcp_558_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 03/11/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Infants should be provided with effective feeding skills with evidence-based care practices to ensure nutritional tolerance and maximize the growth and development in preterm infants. AIM This study aimed to examine the effects of "oral administration of breast milk droplets" and "palatal stimulation with a finger" methods on feed tolerance in preterm newborns fed via an orogastric tube. METHODS This randomized controlled trial was conducted in the neonatal intensive care unit of a private hospital. The study included 90 premature newborns born at the 28th-36th gestational weeks and admitted to intensive care. We applied breast milk droplets inside the oral cavity of newborns (30) in one of the intervention groups and stimulated the palate of newborns (30) by using a finger in the other. We performed these interventions every 3 hours for 5 minutes at the feeding times of the newborns for 7 days. Babies in the control group were not applied any intervention. We used SPSS (Statistical Package for Social Sciences) 22.0 for Windows software for statistical analyses. RESULTS The number of defecations, frequencies of residuals, body weight, and abdominal circumference were significantly different between the groups (P < 0.05). The increments in body weight and reductions in abdominal circumference were significantly different between the groups in the study (P < 0.05). CONCLUSION Palatal stimulation with a finger acted on feed tolerance more favorably than the oral administration of breast milk droplets or no intervention in preterm infants fed via OGT. We suggest primarily that palatal stimulation with a finger and secondarily the oral administration of breast milk droplets as the two methods to be employed to overcome feeding intolerance, which is a significant problem in premature infants.
Collapse
Affiliation(s)
- C Tunç
- Istanbul Medipol University, Institute of Health Science, Istanbul Turkey
| | | |
Collapse
|
23
|
Blesa-Baviera L, Albors A, Samblas P, Maraguat Ú, Coronel-Rodríguez C, Abad B, Viciano E, Pérez-Sádaba FJ, Martínez-Costa C. Growth and gastrointestinal tolerance of healthy formula-fed infants: a multicentre, prospective observational study. BMC Pediatr 2025; 25:229. [PMID: 40128711 PMCID: PMC11934750 DOI: 10.1186/s12887-025-05446-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 01/19/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Infant formula with human milk oligosaccharides (HMOs) and increased β-palmitate mimics breast milk nutritional composition and clinical benefits. We aimed to assess formula-fed infant growth, gastrointestinal tolerance, infections, and parental satisfaction with a partly fermented infant formula with an improved lipid profile (enriched with β-palmitate and docosahexaenoic/arachidonic acid) and short and long-chain oligosaccharides (scGOS/lcFOS [9:1]) and HMOs. METHODS A prospective descriptive observational study in healthy infants with formula feeding or breastfeeding (reference population) was conducted in six Spanish primary care centres following routine clinical practice. In the first, second and fourth month of life visits sociodemographic, clinical, and anthropometric variables (weight, length, head circumference), stool consistency (Brussels Infant and Toddler Stool Scale [BITSS]), gastrointestinal symptoms, infections incidence and associated healthcare resource utilisation, and caregivers' satisfaction with formula were collected. A descriptive statistical analysis was performed (STATA-v.14). Growth was estimated as the mean (standard deviation) increase in the anthropometric variables and z-scores. RESULTS A total of 61 formula-fed and 65 breastfed infants were included in the study (50.8% male). The average increase in weight, length and head circumference in the formula feeding and in the breastfeeding groups from the first to the fourth month of life was 2,566 (496) g, 9.7 (1.7) cm and 4.4 (1.0) cm, and 2,571 (702) g, 9.8 (1.8) cm and 4.4 (1.1) cm, respectively. The weight z-score was -0.1 (0.7) for formula-fed and 0.1 (1.1) for breastfed infants. In all visits, more than 88% of infants had loose/watery stools and most infants suffered gastrointestinal symptoms with low/medium frequency. In the fourth month of life visit, 16 (26.2%) formula-fed and 16 (24.6%) breastfed infants had infections, mainly respiratory, with 16% of formula-fed and 12% of breastfed infants requiring treatment. Most formula-feeding caregivers had a good/very good opinion of formula (85.2%). 75.4% infants drank the whole feeding bottle. CONCLUSIONS The growth, gastrointestinal tolerance, and incidence of infections of healthy formula-fed infants during the first four months of life were appropriate and in line with WHO standards. Formula feeding caregivers were satisfied with this partly fermented infant formula with an improved lipid profile and oligosaccharides.
Collapse
Affiliation(s)
| | - Ana Albors
- Paediatrician. Primary Health Care Centre Trafalgar, Valencia, Spain
| | - Pedro Samblas
- Paediatrician. Primary Health Care Centre El Restón, Madrid, Spain
| | - Úrsula Maraguat
- Paediatrician. Primary Health Care Centre Serrería I, Valencia, Spain
| | | | - Beatriz Abad
- Paediatrician. Primary Health Care Centre Malvarrosa, Valencia, Spain
| | - Elena Viciano
- Outcomes'10 (a ProductLife Group Company), Castellón, Spain
| | | | - Cecilia Martínez-Costa
- Department of Paediatrics, University of Valencia, INCLIVA Biomedical Research Institute, Hospital Clínico Universitario de Valencia, Valencia, Spain.
| |
Collapse
|
24
|
Suri J, Gilmour R. Expediting Glycospace Exploration: Therapeutic Glycans via Automated Synthesis. Angew Chem Int Ed Engl 2025; 64:e202422766. [PMID: 39936247 PMCID: PMC11933530 DOI: 10.1002/anie.202422766] [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: 11/22/2024] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 02/13/2025]
Abstract
Glycans regulate a vast spectrum of disease-related processes, yet effectively leveraging these important mediators in a therapeutic context remains a frontier in contemporary medicine. Unlike many other classes of clinically important biopolymers, carbohydrates derive from discrete biosynthetic pathways and are not produced directly from genes. The conspicuous absence of a biological blueprint to achieve amplification creates a persistent challenge in obtaining well-defined glycostructures for therapeutic translation. Isolating purified sugars from biological sources is not without challenge, rendering synthetic organic chemistry the nexus of this advancing field. Chemical synthesis has proven to be an unfaltering pillar in the production of complex glycans, but laborious syntheses coupled with purification challenges frequently introduce reproducibility issues. In an effort to reconcile these preparative challenges with the societal importance of glycans, automated glycan synthesis was conceptualised at the start of the 21st century. This rapidly expanding, multifaceted field of scientific endeavor has effectively merged synthetic chemistry with technology and engineering to expedite the precision synthesis of target glycans. This minireview describes the structural diversity and function of glycans generated by automated glycan synthesis platforms over the last five years. The translational impact of these advances is discussed together with current limitations and future directions.
Collapse
Affiliation(s)
- James Suri
- Institute for Organic ChemistryUniversity of MünsterCorrensstraße 3648149MünsterGermany
- Cells in Motion (CiM) Interfaculty CenterRöntgenstraße 16D-48149MünsterGermany
| | - Ryan Gilmour
- Institute for Organic ChemistryUniversity of MünsterCorrensstraße 3648149MünsterGermany
- Cells in Motion (CiM) Interfaculty CenterRöntgenstraße 16D-48149MünsterGermany
| |
Collapse
|
25
|
Liao J, Wang M, Li H, Li T, Deng Z, Li J, Zheng L, Yan Y, Duan S, Zhang B. Human Milk Oligosaccharide LNnT Promotes Intestinal Epithelial Growth and Maturation During the Early Life of Infant Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:6678-6690. [PMID: 40048505 DOI: 10.1021/acs.jafc.4c10055] [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: 03/20/2025]
Abstract
Lacto-N-neotetraose (LNnT) is a prevalent neutral core human milk oligosaccharides (HMOs) recognized for its numerous benefits to infant health. In infant formula, galactooligosaccharide (GOS) are frequently used as substitutes for HMOs. However, the regulatory roles of LNnT and GOS in early intestinal development are not yet fully understood. This study aims to elucidate the effects of LNnT and GOS on intestinal development during early life. Our findings show that administering LNnT or GOS significantly increased the spleen and liver indices of infant mice at postnatal day 21. Immunofluorescence and qPCR analysis showed that feeding LNnT significantly promoted the proliferation and differentiation of intestinal stem cells (ISCs) in the colon of infant mice at postnatal day 21, and increased the expression of differentiation markers of goblet cells, intestinal epithelial cells, Paneth cells, and intestinal endocrine cells. Conversely, feeding GOS had no significant effect on the proliferation and differentiation of ISCs. Furthermore, intestinal microbiota analysis showed that LNnT increased the microbiota associated with intestinal regeneration and ISCs proliferation and differentiation in infant mice at postnatal day 21. In conclusion, LNnT promoted ISCs proliferation and differentiation in the colon and alters the composition and function of the intestinal microbiota to support intestinal development in infant mice.
Collapse
Affiliation(s)
- Jinqiang Liao
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047 Jiangxi, China
| | - Minghui Wang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047 Jiangxi, China
| | - Hongyan Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047 Jiangxi, China
- International Institute of Food Innovation, Nanchang University, Nanchang 330051 Jiangxi, China
| | - Ting Li
- Yili Maternal and Infant Nutrition Institute (YMINI), Inner Mongolia Yili Industrial Group, Co. Ltd, Beijing 100070, China
- Inner Mongolia Dairy Technology Research Institute Co. Ltd, Hohhot 010110, China
| | - Zeyuan Deng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047 Jiangxi, China
- International Institute of Food Innovation, Nanchang University, Nanchang 330051 Jiangxi, China
| | - Jing Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047 Jiangxi, China
- International Institute of Food Innovation, Nanchang University, Nanchang 330051 Jiangxi, China
| | - Liufeng Zheng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047 Jiangxi, China
- International Institute of Food Innovation, Nanchang University, Nanchang 330051 Jiangxi, China
| | - Yalu Yan
- Yili Maternal and Infant Nutrition Institute (YMINI), Inner Mongolia Yili Industrial Group, Co. Ltd, Beijing 100070, China
- Inner Mongolia Dairy Technology Research Institute Co. Ltd, Hohhot 010110, China
| | - Sufang Duan
- Yili Maternal and Infant Nutrition Institute (YMINI), Inner Mongolia Yili Industrial Group, Co. Ltd, Beijing 100070, China
- Inner Mongolia Dairy Technology Research Institute Co. Ltd, Hohhot 010110, China
| | - Bing Zhang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047 Jiangxi, China
- International Institute of Food Innovation, Nanchang University, Nanchang 330051 Jiangxi, China
| |
Collapse
|
26
|
Nyquist SK, Annepureddy LD, Sejane K, Furst A, Trahan GD, Rudolph MC, Twigger AJ, Bode L, Engelhardt BE, Martin Carli JF, Goods BA. Integrated 'omics analysis reveals human milk oligosaccharide biosynthesis programs in human lactocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.17.643803. [PMID: 40166160 PMCID: PMC11956926 DOI: 10.1101/2025.03.17.643803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Human milk oligosaccharides (HMOs) are integral to infant health. Yet, their complex biosynthesis pathways in the mammary gland during lactation remain under characterized. To address this knowledge gap, we performed integrated analyses of single-cell RNA-sequencing (scRNA-seq) datasets combined with select HMO concentration measures. We identify differential expression patterns of known HMO synthesis genes in epithelial subsets and nominate several candidate genes that vary with HMO concentration. Additionally, we identify novel gene patterns and transcription factors that may regulate the expression of HMO biosynthesis genes and the cellular pathways supporting HMO production. Finally, we demonstrate that co-expression of HMO synthesis genes and milk fat synthesis genes is limited, suggesting distinct epithelial cell subtypes may be responsible for the production of different milk components. Our study suggests that HMO synthesis may be achieved through cell type specialization within the lactocyte compartment.
Collapse
|
27
|
Gass DT, Pritchard AM, Alberti SN, Gallagher ES. Electron Transfer Higher-Energy Collisional Dissociation Can Distinguish Cobalt-Adducted Isomers of Human Milk Oligosaccharides. Anal Chem 2025; 97:5507-5516. [PMID: 40035718 DOI: 10.1021/acs.analchem.4c05405] [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: 03/06/2025]
Abstract
The ability to identify isomers of human milk oligosaccharides (HMOs) remains a challenge. Typically, HMOs are analyzed using a combination of liquid chromatography and tandem mass spectrometry (LC-MS/MS). Yet, separation methods have been unable to resolve several HMO isomers, necessitating the need for fragmentation methods that can differentiate these structures within mixtures. Common fragmentation methods, such as collision-induced dissociation (CID), often fail to form fragments that can differentiate the linkage and branching isomers. Previously, we determined that electron transfer dissociation (ETD) yields distinct fragmentation products when using certain metal-charge carriers, such as Co2+. Here, we compare fragmentation methods, including CID, higher-energy collisional dissociation (HCD), ETD, and electron transfer, higher-energy collisional dissociation (EThcD) for Co2+- and Na+-adducted HMO isomers. The formation of several fragments using only ETD and EThcD, but not CID and HCD, indicates that certain fragmentation products only form from electron dissociation. EThcD enabled differentiation of Co2+-adducted tri-, tetra-, and pentasaccharide isomers, while CID, HCD, and ETD were incapable of differentiating all the examined HMO isomers as Co2+, Na+, and 2Na2+ adducts. We also show that Co2+ adduction combined with EThcD generates fragments that are related to the core structure and fucose branching location, allowing for further validation of the HMO structures. This work establishes a general method using fragmentation that can be used to differentiate HMOs, which should enable improved identification and structural validation of various carbohydrate linkage and branching isomers.
Collapse
Affiliation(s)
- Darren T Gass
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76798, United States
| | - Andrew M Pritchard
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76798, United States
| | - Sebastian N Alberti
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76798, United States
| | - Elyssia S Gallagher
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76798, United States
| |
Collapse
|
28
|
Gonsalves J, Bauzá-Martinez J, Stahl B, Dingess KA, Mank M. Robust and High-Resolution All-Ion Fragmentation LC-ESI-IM-MS Analysis for In-Depth Characterization or Profiling of Up to 200 Human Milk Oligosaccharides. Anal Chem 2025; 97:5563-5574. [PMID: 40047520 PMCID: PMC11923967 DOI: 10.1021/acs.analchem.4c06081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/28/2025] [Accepted: 02/25/2025] [Indexed: 03/19/2025]
Abstract
Human milk oligosaccharides (HMOs) represent the third most abundant fraction of biomolecules in human milk (HM) and play a crucial role in infant health and development. The unique contributions of HMOs to healthy development of breast-fed infants are assumed to rely on the extraordinary complexity and diversity of HMO isomeric structures, which in turn still cause a huge analytical challenge. Many contemporary analytical methods aiming for more detailed HMO characterization combine ion mobility (IM) with LC-MS for enhanced structural resolution but are typically lacking the robustness necessary for application to HM cohorts with hundreds of samples. To overcome these challenges, we introduce a novel, robust all-ion fragmentation (AIF) LC-ESI-IM-MS method integrating four analytical dimensions: high-resolution LC separation, IM drift time, accurate mass precursor, and fragment ion measurements. This four-dimensional (4D) analytical characterization is sufficient for resolving various HMO structural isomers in an efficient way. Thereby, up to 200 HMO compounds with a maximum degree of polymerization of 13 could be simultaneously identified and relatively quantified. We devised two methods using this 4D analytical approach. One intended for in-depth characterization of multiple known but also novel HMO structures and the second is designed for robust, increased-throughput analyses. With the first approach, five trifucosyl-lacto-N-tetraose isomers (TF-LNTs), four of which were never detected before in HM, as well as additional difucosyl-lacto-N-heaose isomers (DF-LNHs), were revealed and structures fully elucidated by AIF and IM. This exemplifies the potential of our method for in-depth characterization of novel complex HMO structures. Furthermore, the increased-throughput method featuring a shorter LC gradient was applied to real-world HM samples. Here, we could differentiate the HM types I-IV based on a broader range of partly new marker HMOs. We could also derive valuable new insights into variations of multiple and rare HMOs up to DP 11 across lactational stages. Overall, our AIF LC-ESI-IM-MS approach facilitates in-depth monitoring and confident identification of a broad array of distinct and simple to very complex HMOs. We envision this robust AIF LC-ESI-IM-MS approach to advance HMO research by facilitating the characterization of a broad range of HMOs in high numbers of HM samples. This may help to further extend our understanding about HMOs structure-function relationships relevant for infants' healthy development.
Collapse
Affiliation(s)
- John Gonsalves
- Danone Research
& Innovation, Uppsalalaan
12, 3584 CT Utrecht, The Netherlands
| | | | - Bernd Stahl
- Danone Research
& Innovation, Uppsalalaan
12, 3584 CT Utrecht, The Netherlands
- Utrecht Institute
for Pharmaceutical Sciences, Department of Chemical Biology &
Drug Discovery, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Kelly A. Dingess
- Danone Research
& Innovation, Uppsalalaan
12, 3584 CT Utrecht, The Netherlands
| | - Marko Mank
- Danone Research
& Innovation, Uppsalalaan
12, 3584 CT Utrecht, The Netherlands
| |
Collapse
|
29
|
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
|
30
|
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
|
31
|
Sinha R, Ottosen EN, Ngwaga T, Shames SR, DiRita VJ. Carbapenem-resistant Enterobacter hormaechei uses mucus metabolism to facilitate gastrointestinal colonization. mBio 2025; 16:e0288424. [PMID: 39878485 PMCID: PMC11898723 DOI: 10.1128/mbio.02884-24] [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: 09/24/2024] [Accepted: 01/06/2025] [Indexed: 01/31/2025] Open
Abstract
The emergence and global spread of carbapenem-resistant Enterobacter cloacae complex species present a pressing public health challenge. Carbapenem-resistant Enterobacter spp. cause a wide variety of infections, including septic shock fatalities in newborns and immunocompromised adults. The intestine may be a major reservoir for these resistant strains, either by facilitating contamination of fomites and transfer to susceptible individuals, or through translocation from the gut to the bloodstream. For this reason, we sought to establish a neonatal mouse model to investigate the mechanisms underpinning gut colonization by carbapenem-resistant Enterobacter hormaechei. We describe a new mouse model to study gut colonization by Enterobacter spp., leading to vital insights into the adaptation of carbapenem-resistant E. hormaechei to the gut environment during the early stages of intestinal colonization. We observed successful colonization and proliferation of E. hormaechei in the 5-day-old infant mouse gut, with primary localization to the colon following oral inoculation. We also uncovered evidence that E. hormaechei uses mucus as a carbon source during colonization of the colon. Our findings underscore the importance of oxygen-dependent metabolic pathways, including the pyruvate dehydrogenase complex and N-acetyl-D-glucosamine metabolism, in gut colonization and proliferation, which aligns with previous human studies. These insights are essential for developing novel therapeutic strategies that can serve as decolonization therapies in at-risk populations.IMPORTANCEBloodstream infections caused by Enterobacter spp. pose a significant clinical threat. The intestine acts as the primary site for colonization and serves as a reservoir for infection. To combat this pathogen, it is crucial to understand how carbapenem-resistant Enterobacter spp. colonize the gut, as such knowledge can pave the way for alternative therapeutic targets. In this study, we developed a novel neonatal mouse model for gastrointestinal colonization by Enterobacter spp. and discovered that mucus plays a key role as a carbon source during colonization. Additionally, we identified two mucus catabolism pathways that contribute to intestinal colonization by carbapenem-resistant E. hormaechei. This new mouse model offers valuable insights into host-pathogen interactions and helps identify critical gastrointestinal fitness factors of Enterobacter, potentially guiding the development of vaccines and alternative therapeutic strategies to minimize intestinal carriage in patient populations at risk of infection with Enterobacter spp.
Collapse
Affiliation(s)
- Ritam Sinha
- Department of Microbiology, Genetics, & Immunology, Michigan State University, East Lansing, Michigan, USA
| | - Elizabeth N. Ottosen
- Department of Microbiology, Genetics, & Immunology, Michigan State University, East Lansing, Michigan, USA
| | | | - Stephanie R. Shames
- Department of Microbiology, Genetics, & Immunology, Michigan State University, East Lansing, Michigan, USA
| | - Victor J. DiRita
- Department of Microbiology, Genetics, & Immunology, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
32
|
Xia Z, Lao C, Wu J, Jin Y, Chen X, Li H, Fan X, Yuan L, Sun L. Optimization of l-Fucose Biosynthesis in Escherichia coli through Pathway Engineering and Mixed Carbon Source Strategy. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:6102-6112. [PMID: 40029204 DOI: 10.1021/acs.jafc.4c12544] [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: 03/05/2025]
Abstract
This study presents an engineered strain of Escherichia coli specifically designed to enhance the production of l-fucose while minimizing residues of 2'-fucosyllactose. The optimization strategies employed include the selection of key enzymes, optimization of gene copy numbers, and fermentation using mixed carbon sources. The metabolic flux was directed toward l-fucose synthesis by integrating preferred 1,2-fucosyltransferase and α-l-fucosidase into the genome. Furthermore, the gene copy numbers were optimized to enhance enzyme expression, thereby increasing l-fucose production. Additionally, the supply of guanosine 5'-triphosphate was improved, and cofactors were regenerated to better regulate metabolism. Modifications to transporter proteins effectively reduced the accumulation of 2'-fucosyllactose. The implementation of a glucose/glycerol co-fermentation strategy enhanced carbon flux distribution and strain efficiency. The optimized strain achieved a yield of 91.90 g/L of l-fucose in a 5 L bioreactor, representing an 80.01% increase over previous yields, with a productivity of 1.18 g L-1 h-1. This yield is the highest reported for l-fucose, demonstrating its potential for industrial production.
Collapse
Affiliation(s)
- Zihan Xia
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China
- Institute of Plasma Physics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China
| | - Caiwen Lao
- Hefei CAS Health Bio-Industrial Technology Institute Company, Limited, Hefei, Anhui 230031, China
| | - Jinyong Wu
- Institute of Plasma Physics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China
| | - Yiwen Jin
- Hefei CAS Health Bio-Industrial Technology Institute Company, Limited, Hefei, Anhui 230031, China
| | - Xiangsong Chen
- Institute of Plasma Physics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China
| | - He Li
- Hefei CAS Health Bio-Industrial Technology Institute Company, Limited, Hefei, Anhui 230031, China
| | - Xijie Fan
- Hefei CAS Health Bio-Industrial Technology Institute Company, Limited, Hefei, Anhui 230031, China
| | - Lixia Yuan
- Institute of Plasma Physics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China
| | - Lijie Sun
- Institute of Plasma Physics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China
| |
Collapse
|
33
|
Lazarini T, Tonon KM, de Araujo Filho HB, de Morais MB. Bifidogenic Effect of 2'-Fucosyllactose (2'-FL) on the Gut Microbiome of Healthy Formula-Fed Infants: A Randomized Clinical Trial. Nutrients 2025; 17:973. [PMID: 40290019 PMCID: PMC11944528 DOI: 10.3390/nu17060973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 02/28/2025] [Accepted: 03/05/2025] [Indexed: 04/30/2025] Open
Abstract
Breast milk is rich in bioactive components, especially human milk oligosaccharides (HMOs), which are crucial for establishing gut microbiota. The 2'-FL (2-Fucosyllactose), one of the most abundant oligosaccharides in breast milk, functions as a selective prebiotic. Objective: To examine the effect of adding 2'-FL (2-Fucosyllactose) to an infant formula containing prebiotic galacto-oligosaccharides (GOSs) and fructo-oligosaccharides (FOSs) on the gut microbiome of healthy formula-fed infants. Methods: This study enrolled infants from three groups: an HMO experimental group (n = 29), a GOS/FOS control group (n = 30), and an exclusively breastfed (breast milk [BM]) reference group (n = 28). Fecal samples from the three groups in the first and fourth months of life were analyzed. The V3 and V4 regions of the 16S rRNA gene were amplified and sequenced on the Illumina MiSeq. ANOVA, Kruskal-Wallis, richness indices (Chao1, Shannon), UniFrac distances, and the Adonis tests were used to perform statistical analyses on the relative abundance of phyla and genera, as well as the alpha and beta-diversity of the gut microbiota. Results: After intervention, Actinobacteriota emerged as the predominant phylum in both the HMO (60.4%) and BM (46.6%) groups. Bifidobacterium and Escherichia-Shigella were identified as the two most abundant bacterial genera in both groups. Nevertheless, the statistical analysis showed that the relative abundance of Bifidobacterium in the HMO formula-fed group after intervention was similar to that in the BM group (p > 0.05). Infants in the HMO and GOS/FOS groups showed higher relative abundance of [Ruminococcus]_gnavus_group bacteria compared to those in the BM group. Groups fed with infant formula demonstrated higher alpha-diversity of gut microbiota compared to breastfed infants (p < 0.05), at the time of admission as well as after the intervention. Beta-diversity was significantly different among the three groups, according to type of feeding. Infants fed a 2'-FL-supplemented infant formula exhibited growth comparable to that of breastfed infants throughout the intervention period, demonstrating that the formula was both safe and well tolerated. Conclusions: Adding 2'-FL to an infant formula containing 4 g/L of GOS + FOS resulted in a stronger bifidogenic effect compared to the formula without 2'-FL.
Collapse
Affiliation(s)
- Tamara Lazarini
- Nutrition Postgraduate Program, Universidade Federal de São Paulo, São Paulo 04023-062, Brazil;
| | - Karina Merini Tonon
- Department of Environmental & Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA;
| | | | - Mauro Batista de Morais
- Nutrition Postgraduate Program, Universidade Federal de São Paulo, São Paulo 04023-062, Brazil;
- Division of Pediatric Gastroenterology, Universidade Federal de São Paulo, São Paulo 04023-062, Brazil;
| |
Collapse
|
34
|
Media TS, Ramesh M, Lee OI, Ubaka LN, Harn DA, Norberg T, Quinn F, Garg A. The Human Milk Oligosaccharide Lacto-N-Fucopentaose III Conjugated to Dextran Inhibits HIV Replication in Primary Human Macrophages. Nutrients 2025; 17:890. [PMID: 40077760 PMCID: PMC11901455 DOI: 10.3390/nu17050890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/20/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Background/Objectives: Individuals with HIV on combined antiretroviral therapy (ART) with virologic suppression exhibit chronic immune activation and immune dysfunction. Numerous studies have shown that human milk oligosaccharide (HMO) controls the postnatal transmission of HIV-1, but its effect on adult HIV-1 infection is not known. The purpose of this study was to investigate the anti-HIV activity of Lacto-N-fucopentaose III (LNFPIII) in adult blood-borne macrophages. Methods: Primary human monocyte-derived macrophages from the blood of HIV-seronegative individuals were infected with HIV and treated with or without dextran-conjugated LNFPIII (P3DEX). HIV replication was measured by quantifying the accumulation of HIV Gag p24 in the culture supernatants by ELISA. The quantities of chemokines MIP-1α, MIP-1β, and CCL5 in the culture supernatant were also measured by ELISA. The expression of IL-1β, IL-18, TNFα, IL-10, BECN1, and housekeeping gene HuPO in the macrophages was determined by qRT PCR. The expression of NF-kB, LC3, p62, and β-actin was measured by immunoblotting. Results: We found that P3DEX controls HIV replication without affecting HIV binding and/or internalization by human macrophages. The treatment of HIV-infected macrophages with P3DEX increased the quantity of beta (β)-chemokines MIP-1α, CCL5, and MIP-1β, which are known to have anti-HIV activity. Furthermore, the treatment of HIV-infected macrophages with P3DEX increased autophagic flux in a TLR8-dependent manner and ameliorated the expression of proinflammatory cytokines. These results suggest that P3DEX is a prominent milk-derived sugar that simultaneously augments anti-viral mechanisms and controls immune activation. These findings prudently justify the use and clinical development of P3DEX as a host-directed therapeutic option for people living with HIV.
Collapse
Affiliation(s)
- Tablow Shwan Media
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; (T.S.M.); (M.R.); (O.I.L.); (L.N.U.); (D.A.H.); (F.Q.)
| | - Medhini Ramesh
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; (T.S.M.); (M.R.); (O.I.L.); (L.N.U.); (D.A.H.); (F.Q.)
| | - Olivia Isa Lee
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; (T.S.M.); (M.R.); (O.I.L.); (L.N.U.); (D.A.H.); (F.Q.)
| | - Lucy Njideka Ubaka
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; (T.S.M.); (M.R.); (O.I.L.); (L.N.U.); (D.A.H.); (F.Q.)
| | - Donald A. Harn
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; (T.S.M.); (M.R.); (O.I.L.); (L.N.U.); (D.A.H.); (F.Q.)
| | - Thomas Norberg
- Department of Biochemistry-BMC, Uppsala University, 753 10 Uppsala, Sweden;
| | - Frederick Quinn
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; (T.S.M.); (M.R.); (O.I.L.); (L.N.U.); (D.A.H.); (F.Q.)
| | - Ankita Garg
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; (T.S.M.); (M.R.); (O.I.L.); (L.N.U.); (D.A.H.); (F.Q.)
| |
Collapse
|
35
|
Chen Q, Wang X, Zhang P, Li B. Recent trends in human milk oligosaccharides: New synthesis technology, regulatory effects, and mechanisms of non-intestinal functions. Compr Rev Food Sci Food Saf 2025; 24:e70147. [PMID: 40091651 DOI: 10.1111/1541-4337.70147] [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: 10/18/2024] [Revised: 01/30/2025] [Accepted: 02/07/2025] [Indexed: 03/19/2025]
Abstract
Recently, the non-intestinal functions of human milk oligosaccharides (HMOs) have been widely documented, including their roles in promoting brain development and growth, as well as ameliorating anxiety, allergies, and obesity. Understanding their mechanisms of action is becoming increasingly critical. Furthermore, these effects are frequently associated with the type and structure of HMOs. As an innovative technology, "plant factory" is expected to complement traditional synthesis technology. This study reviews the novel "plant factory" synthesis techniques. Particular emphasis is placed on the processes, advantages, and limitations of "plant factory" synthesis of HMOs. This technology can express genes related to HMO synthesis instantaneously in plant leaves, thereby enabling the rapid and cost-effective generation of HMOs. However, "plant factory" technology remains underdeveloped, and challenges related to low yield and unsustainable production must be addressed. Furthermore, we present an overview of the most recent clinical and preclinical studies on the non-intestinal functions of HMOs. This review emphasizes the mechanisms of action underlying the non-intestinal functions of HMOs. HMOs primarily exert non-intestinal functions through the cleavage of beneficial monomer components, metabolism to produce advantageous metabolites, and regulation of immune responses.
Collapse
Affiliation(s)
- Qingxue Chen
- Food College, Northeast Agricultural University, Harbin, China
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Xiangxin Wang
- Food College, Northeast Agricultural University, Harbin, China
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Peng Zhang
- Food College, Northeast Agricultural University, Harbin, China
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Bailiang Li
- Food College, Northeast Agricultural University, Harbin, China
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| |
Collapse
|
36
|
Ahn J, Choi E, Lee KG. Analysis of volatiles and α-dicarbonyl compounds in Maillard reaction products derived from 2'-fucosyllactose and amino acids. Food Res Int 2025; 205:115975. [PMID: 40032468 DOI: 10.1016/j.foodres.2025.115975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 01/12/2025] [Accepted: 02/08/2025] [Indexed: 03/05/2025]
Abstract
This study aims to investigate the volatile and α-dicarbonyl compounds (α-DCs) formed in Maillard reactions between 2'-fucosyllactose (2'-FL) and amino acids, with the goal of exploring their potential as flavoring agents and enhancing food quality and safety. The effects of pH, temperature, reaction time, and amino acid concentration on α-DC production were evaluated. Fucose generated the most α-DCs, whereas 2'-FL produced the least. α-DC formation increased with increasing pH, reaction time, temperature, and amino acid concentration. Among the amino acids evaluated, threonine elicited the highest α-DC production. In total, 50 volatile compounds were identified, with 2'-FL and lactose primarily forming furan and furan derivatives. In particular, 2'-FL yielded greater amounts of 2-furfural, 2-acetylfuran, 5-methylfurfural, furfuryl alcohol, and 2-furanmethanol than other monosaccharides. These findings highlight the potential of 2'-FL as a flavouring agent and enhance our understanding of α-DC formation during food processing and storage.
Collapse
Affiliation(s)
- Junghyun Ahn
- Department of Food Science and Biotechnology, Dongguk University-Seoul, 32, Dongguk-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10326, Republic of Korea
| | - Eunyeong Choi
- Department of Food Science and Biotechnology, Dongguk University-Seoul, 32, Dongguk-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10326, Republic of Korea
| | - Kwang-Geun Lee
- Department of Food Science and Biotechnology, Dongguk University-Seoul, 32, Dongguk-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10326, Republic of Korea.
| |
Collapse
|
37
|
Ren X, Xiao M, Zhang L, Zhang H, Li R, Zhu C, Li D, Fu X, Mou H. Impact of 2'-fucosyllactose on adult gut microbiota composition and metabolome based on colonic fermentation and prebiotic quantitative evaluation. J Food Sci 2025; 90:e70084. [PMID: 40135481 DOI: 10.1111/1750-3841.70084] [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: 11/24/2024] [Revised: 01/23/2025] [Accepted: 02/04/2025] [Indexed: 03/27/2025]
Abstract
2'-Fucosyllactose (2'-FL), a predominant human milk oligosaccharide, is widely employed as a commercial infant prebiotic. Nevertheless, the benefits of 2'-FL on complex gut microbiota need further exploration. The modulation of 2'-FL on adult gut microbiota composition and metabolome, particularly short-chain fatty acids (SCFAs) and tryptophan metabolism, was investigated via colonic fermentation, single bacterial strains cultivation, and prebiotic activity scoring evaluation. The 2'-FL fermentation in a complex microbial community indicated promising effects, including an increase in Bifidobacterium levels and changes in metabolic levels of tryptophan, SCFAs, and vitamins. Correlation analysis of microbiota and metabolome highlighted a positive correlation between Bifidobacterium and metabolites derived from SCFAs and indole. Scoring formulas for bacterial growth and substrate utilization were introduced to compare the overall response of 15 bacterial strains from key genera that were identified by 16S rRNA profiling to several prebiotics. In the cultivation of single bacterial strains, 2'-FL was selectively used by Bifidobacteria infantis and Enterococcus faecalis and was not degraded by potential pathogenic strains. This selective promotion of probiotics by 2'-FL was observed according to the scoring results, as 2'-FL showed higher scores for bacterial growth and substrate utilization when compared to galacto-oligosaccharides and fucose. This study demonstrated beneficial bifidogenic effect of 2'-FL in an adult complex microbial community and the potential of scoring evaluation as a quantitative tool for measuring the prebiotic effects of different oligosaccharides.
Collapse
Affiliation(s)
- Xinmiao Ren
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China
| | - Mengshi Xiao
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China
| | - Ling Zhang
- Qingdao Women and Children's Hospital, Qingdao, Shandong, China
| | - Hang Zhang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong, China
| | - Rong Li
- Qingdao Women and Children's Hospital, Qingdao, Shandong, China
| | - Changliang Zhu
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China
| | - Dongyu Li
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China
| | - Xiaodan Fu
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China
| | - Haijin Mou
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China
| |
Collapse
|
38
|
Younge N, Patel RM. Probiotics and the Risk of Infection. Clin Perinatol 2025; 52:87-100. [PMID: 39892956 PMCID: PMC11789005 DOI: 10.1016/j.clp.2024.10.006] [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] [Indexed: 02/04/2025]
Abstract
Probiotic use has increased in preterm infants and may reduce the risk of necrotizing enterocolitis. Probiotic-associated infection is a concern for infants receiving probiotic supplementation in the neonatal intensive care unit, as highlighted by a recent case and subsequent action by the United States Food and Drug Administration. Based on reports to date, invasive infection is an infrequent but known risk of probiotic supplementation. In this article, we discuss the epidemiology and pathophysiology of invasive infection in preterm infants, review the benefits and risks of probiotic as regulations and available products continue to evolve.
Collapse
Affiliation(s)
- Noelle Younge
- Department of Pediatrics, Duke University, 2400 Pratt Street, DUMC Box 2739, Durham, NC 27705, USA
| | - Ravi M Patel
- Department of Pediatrics, Emory University and Children's Healthcare of Atlanta, Arthur M. Blank Hospital, 2220 North Druid Hills Road NE, CL.06323, Atlanta, GA 30329, USA.
| |
Collapse
|
39
|
Wejryd E, Freiholtz Jern E, Marchini G, Åden U, Landberg E, Abrahamsson T. Human Milk Oligosaccharides in Breast Milk at Two Weeks of Age in Relation to Neurodevelopment in 2-Year-Old Children Born Extremely Preterm: An Explorative Trial. Nutrients 2025; 17:832. [PMID: 40077703 PMCID: PMC11902041 DOI: 10.3390/nu17050832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Background: Preventing neurodevelopmental impairment after extremely preterm birth remains challenging. While breast milk feeding is linked to better neurodevelopment, the underlying mechanisms are unclear. This study explored the association between individual human milk oligosaccharides (HMO) and neurodevelopment at two years of age in extremely preterm children. Methods: Milk samples from mothers of 76 extremely preterm infants collected at two weeks after birth were analyzed for 15 dominant HMOs. Register data from examination and Bayley-III neurodevelopmental assessment at two years' corrected age was retrieved and categorized into levels of impairment. An exploratory analysis examined associations between the HMO composition and neurodevelopment. Results: Bioinformatic volcano plots revealed associations between specific HMOs and outcomes: 3FL with less neurodevelopmental impairment, LSTb with higher Bayley-III cognitive scores, and LSTa with worse neurodevelopmental impairment outcomes. Spearman correlations indicated LSTa was linked to more neurodevelopmental impairment (p = 0.018), lower language (p = 0.009), and motor (p = 0.02) scores, whereas 3FL correlated with less neurodevelopmental impairment (p = 0.02). Dichotomized analysis showed LSTa was associated with more neurodevelopmental impairment and lower language scores (p < 0.05), 3FL with milder neurodevelopmental impairment (p < 0.05), and LSTb with better cognitive (p < 0.01) and language (p < 0.05) scores. No significant associations were found for HMO diversity, total sialic acid content, or secretor/Lewis patterns. Conclusions: In this explorative hypothesis-generating study, certain HMOs appeared to be associated with both potentially beneficial and adverse neurodevelopmental outcomes in extremely preterm infants. However, these findings should be interpreted with caution, as they do not constitute evidence but rather serve as a preliminary foundation for future hypothesis-driven research.
Collapse
Affiliation(s)
- Erik Wejryd
- Division of Children’s and Women’s Health, Department of Biomedical and Clinical Sciences, Linköping University, 581 83 Linköping, Sweden; (E.W.); (E.F.J.)
- Department of Pediatrics, Vrinnevi Hospital, 601 82 Norrköping, Sweden
| | - Erik Freiholtz Jern
- Division of Children’s and Women’s Health, Department of Biomedical and Clinical Sciences, Linköping University, 581 83 Linköping, Sweden; (E.W.); (E.F.J.)
| | - Giovanna Marchini
- Department of Neonatology, Astrid Lindgren Children’s Hospital, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Ulrika Åden
- Division of Children’s and Women’s Health, Department of Biomedical and Clinical Sciences, Linköping University, 581 83 Linköping, Sweden; (E.W.); (E.F.J.)
- Department of Neonatology, Astrid Lindgren Children’s Hospital, Karolinska University Hospital, 171 76 Stockholm, Sweden
- Department of Women’s and Children’s Health, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Eva Landberg
- Department of Clinical Chemistry and Department of Biomedical and Clinical Sciences, Linköping University, 581 83 Linköping, Sweden;
| | - Thomas Abrahamsson
- Division of Children’s and Women’s Health, Department of Biomedical and Clinical Sciences, Linköping University, 581 83 Linköping, Sweden; (E.W.); (E.F.J.)
- Crown Princess Victoria Children’s Hospital, Linköping University Hospital, 581 85 Linköping, Sweden
| |
Collapse
|
40
|
Patil K, Ayyar BV, Hayes NM, Neill FH, Bode L, Estes MK, Atmar RL, Ramani S. 2'-Fucosyllactose inhibits human norovirus replication in human intestinal enteroids. J Virol 2025; 99:e0093824. [PMID: 39791912 PMCID: PMC11853015 DOI: 10.1128/jvi.00938-24] [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: 05/29/2024] [Accepted: 12/08/2024] [Indexed: 01/12/2025] Open
Abstract
Human noroviruses (HuNoVs) are the leading cause of acute gastroenteritis worldwide. Currently, there are no targeted antivirals for the treatment of HuNoV infection. Histo-blood group antigens (HBGAs) on the intestinal epithelium are cellular attachment factors for HuNoVs; molecules that block the binding of HuNoVs to HBGAs thus have the potential to be developed as antivirals. Human milk oligosaccharides (HMOs) are glycans in human milk with structures analogous to HBGAs. HMOs have been shown to act as decoy receptors to prevent the attachment of multiple enteric pathogens to host cells. Previous X-ray crystallography studies have demonstrated the binding of HMO 2'-fucosyllactose (2'FL) in the same pocket as HBGAs for some HuNoV strains. We evaluated the effect of 2'FL on the replication of a globally dominant GII.4 Sydney [P16] HuNoV strain using human intestinal enteroids (HIEs) from adults and children. A significant reduction in GII.4 Sydney [P16] replication was seen in duodenal and jejunal HIEs from multiple adult donors, all segments of the small intestine from an adult organ donor, and in two pediatric duodenal HIEs. However, 2'FL did not inhibit HuNoV replication in two infant jejunal HIEs that had significantly lower expression of α1-2-fucosylated glycans. 2'FL can be synthesized in large scale, and safety and tolerance have been assessed previously. Our data suggest that 2'FL has the potential to be developed as a therapeutic for HuNoV gastroenteritis. IMPORTANCE Human noroviruses infect the gastrointestinal tract and are a leading cause of acute gastroenteritis worldwide. Common symptoms of norovirus include diarrhea, vomiting, and stomach cramps. Virus shedding and symptoms are prolonged and debilitating in immunocompromised patients. Currently, there are no approved vaccines or targeted antivirals for treating human norovirus infection. Human intestinal enteroids derived from intestinal stem cells allow the successful replication of norovirus in the laboratory and can be used as a physiologically relevant model system to evaluate antivirals. We discovered that 2'-fucosyllactose (2'FL), an oligosaccharide naturally occurring in human milk, inhibits GII.4 norovirus replication in HIEs from multiple donors and thus has the potential to be developed as a therapeutic for human norovirus. These findings have high translational potential since 2'FL from several manufacturers has a "generally recognized as safe" status and can be synthesized on a large scale for immediate application.
Collapse
Affiliation(s)
- Ketki Patil
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - B. Vijayalakshmi Ayyar
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Nicole M. Hayes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Frederick H. Neill
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Lars Bode
- Department of Pediatrics, Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (MOMI CORE), and the Human Milk Institute (HMI), University of California San Diego, La Jolla, California, USA
| | - Mary K. Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Robert L. Atmar
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Sasirekha Ramani
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
41
|
Ghith A, Maleki R, Grzeskowiak LE, Amir LH, Ingman WV. Challenges and Opportunities in Quantifying Bioactive Compounds in Human Breastmilk. Biomolecules 2025; 15:325. [PMID: 40149861 PMCID: PMC11940641 DOI: 10.3390/biom15030325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/29/2025] Open
Abstract
Breastmilk is a complex biological fluid containing over a thousand bioactive proteins, lipids, cells and small molecules that provide nutrition and immunological protection for infants and children. The composition of breastmilk is unique to each individual and can also vary within individuals according to breastfeeding duration, maternal health, time of day, and other factors. As such, the composition of breastmilk can be considered a "fingerprint" that could be interrogated to identify biomarkers of breast health and disease. However, accurate quantification of bioactive components in breastmilk remains a significant challenge. Approaches such as immunoassays and mass spectrometry have been largely applied to study blood or other biological fluids and require validation and optimisation before these techniques can be used to accurately quantify bioactive compounds in breastmilk. Development of protocols specific to breastmilk should be carried out with high precision, confidence, and sensitivity. This review explores the challenges and opportunities associated with different techniques for quantification of breastmilk bioactive components.
Collapse
Affiliation(s)
- Amna Ghith
- Discipline of Surgical Specialties, Adelaide Medical School, University of Adelaide, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia; (A.G.); (R.M.)
- Robinson Research Institute, University of Adelaide, Adelaide, SA 5006, Australia
| | - Reza Maleki
- Discipline of Surgical Specialties, Adelaide Medical School, University of Adelaide, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia; (A.G.); (R.M.)
- Robinson Research Institute, University of Adelaide, Adelaide, SA 5006, Australia
| | - Luke E. Grzeskowiak
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA 5042, Australia;
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, Women’s and Children’s Hospital, North Adelaide, SA 5006, Australia
| | - Lisa H. Amir
- Judith Lumley Centre, School of Nursing and Midwifery, La Trobe University, Bundoora, VIC 3086, Australia;
- Breastfeeding Service, The Royal Women’s Hospital, Parkville, VIC 3050, Australia
| | - Wendy V. Ingman
- Discipline of Surgical Specialties, Adelaide Medical School, University of Adelaide, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia; (A.G.); (R.M.)
- Robinson Research Institute, University of Adelaide, Adelaide, SA 5006, Australia
| |
Collapse
|
42
|
Tseng HK, Lee TY, Chiang YC, Kuo WH, Tseng HW, Wang HK, Ni CK, Lin CC. Versatile Strategy for the Chemoenzymatic Synthesis of Branched Human Milk Oligosaccharides Containing the Lacto-N-Biose Motif. Angew Chem Int Ed Engl 2025; 64:e202419021. [PMID: 39589188 DOI: 10.1002/anie.202419021] [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: 10/02/2024] [Revised: 11/19/2024] [Accepted: 11/26/2024] [Indexed: 11/27/2024]
Abstract
Human milk oligosaccharides (HMOs) exhibit prebiotic, antimicrobial, and immunomodulatory properties and confer significant benefits to infants. Branched HMOs are constructed through diverse glycosidic linkages and prominently feature the lacto-N-biose (LNB, Gal-β1,3-GlcNAc) motif with fucose and/or sialic acid modifications, displaying structural complexity that surpasses that of N- and O-glycans. However, synthesizing comprehensive libraries of branched HMO is challenging due to this complexity. Although a few systematic synthetic strategies have emerged, many of them rely on labor-intensive chemical methodologies or exploit the substrate specificity of human N-acetylglucosaminyltransferase 2 (hGCNT2). In this study, we capitalized on the substrate promiscuities of hGCNT2 and bacterial glycosyltransferases (GTs) to construct a universal tetrasaccharide core in a highly efficient manner. This core was systematically and flexibly extended to generate diverse branched HMOs utilizing the promiscuity of bacterial GTs coupled with N-trifluoroacetyl glucosamine (GlcNTFA), which facilitated sugar chain elongation. The GlcNTFA residues were subsequently converted into various N-modified glucosamines through straightforward chemical manipulations to modulate the activities of additional GTs during glycan extension. These masked amino groups were ultimately reverted to N-acetyl groups, facilitating the synthesis of a broad range of asymmetric and multiantennary HMOs featuring LNB moieties, including many previously inaccessible structures.
Collapse
Affiliation(s)
- Hsin-Kai Tseng
- Department of Chemistry, National Tsing Hua University, 101 Section 2, Kuang Fu Road, Hsinchu, 30013, Taiwan
| | - Ting-Yi Lee
- Department of Chemistry, National Tsing Hua University, 101 Section 2, Kuang Fu Road, Hsinchu, 30013, Taiwan
| | - Yu-Ching Chiang
- Department of Chemistry, National Tsing Hua University, 101 Section 2, Kuang Fu Road, Hsinchu, 30013, Taiwan
| | - Wen-Hua Kuo
- Department of Chemistry, National Tsing Hua University, 101 Section 2, Kuang Fu Road, Hsinchu, 30013, Taiwan
| | - Hsien-Wei Tseng
- Department of Chemistry, National Tsing Hua University, 101 Section 2, Kuang Fu Road, Hsinchu, 30013, Taiwan
| | - Hung-Kai Wang
- Department of Chemistry, National Tsing Hua University, 101 Section 2, Kuang Fu Road, Hsinchu, 30013, Taiwan
| | - Chi-Kung Ni
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 10617, Taiwan
| | - Chun-Cheng Lin
- Department of Chemistry, National Tsing Hua University, 101 Section 2, Kuang Fu Road, Hsinchu, 30013, Taiwan
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| |
Collapse
|
43
|
Renwick S, Furst A, Knip M, Bode L, Danska JS, Allen-Vercoe E. Modulating the developing gut microbiota with 2'-fucosyllactose and pooled human milk oligosaccharides. MICROBIOME 2025; 13:44. [PMID: 39920790 PMCID: PMC11803978 DOI: 10.1186/s40168-025-02034-9] [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: 05/30/2024] [Accepted: 01/08/2025] [Indexed: 02/09/2025]
Abstract
BACKGROUND Synthetic human milk oligosaccharides (HMOs) are used to supplement infant formula despite limited understanding of their impact on the post-weaned developing gut microbiota. Here, we assess the influence of 0.5 g/L 2-fucosyllactose (2'FL) and 4.0 g/L pooled HMOs (pHMOs) on the composition and activity of cultured fecal-derived microbial communities from seven healthy young children. RESULTS Exposure to pHMOs induced significant shifts in both the microbial community composition and metabolic output, including an increased abundance of several genera, notably Bacteroides, and the production of health-associated metabolites. In contrast, 2'FL alone did not lead to substantial changes in the communities. A total of 330 bacterial isolates, spanning 157 species, were cultured from these communities and individually evaluated for their responses to HMOs. Over 100 non-Bifidobacterium species showed enhanced growth upon pHMOs treatment and a high degree of intraspecies variation in HMO metabolism was observed. CONCLUSION Our study provides valuable insight into the health-enhancing properties of HMOs while highlighting the need for future research into the efficacy of incorporating individual structures into infant formula, particularly when aiming to modulate the gut microbiota. Video Abstract.
Collapse
Affiliation(s)
- Simone Renwick
- Family Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (MOMI CORE), the Human Milk Institute (HMI), Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Annalee Furst
- Family Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (MOMI CORE), the Human Milk Institute (HMI), Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Mikael Knip
- Research Programs Unit, Medical Faculty, University of Helsinki, Helsinki, Finland
- New Children's Hospital, Helsinki University Hospital, Helsinki, Finland
- Tampere Center for Child Health Research, Tampere University Hospital, Tampere, Finland
| | - Lars Bode
- Family Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (MOMI CORE), the Human Milk Institute (HMI), Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Jayne S Danska
- The Hospital for Sick Children, Toronto, ON, Canada
- Department of Immunology and Department of Medicine Biophysics, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Emma Allen-Vercoe
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of Guelph, Guelph, ON, Canada.
| |
Collapse
|
44
|
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
|
45
|
Isolauri E, Laitinen K. Resilience to Global Health Challenges Through Nutritional Gut Microbiome Modulation. Nutrients 2025; 17:396. [PMID: 39940253 PMCID: PMC11821120 DOI: 10.3390/nu17030396] [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: 12/20/2024] [Revised: 01/14/2025] [Accepted: 01/20/2025] [Indexed: 02/14/2025] Open
Abstract
As the world faces an escalating challenge of non-communicable diseases (NCDs), with phenotypes ranging from allergic chronic immuno-inflammatory diseases to neuropsychiatric disorders, it becomes evident that their seeds are sown during the early stages of life. Furthermore, within only a few decades, human obesity has reached epidemic proportions and now represents the most serious public health challenge of our time. Recent demonstrations that a growing number of these conditions are linked to aberrant gut microbiota composition and function have evoked active scientific interest in host-microbe crosstalk, characterizing and modulating the gut microbiota in at-risk circumstances. These efforts appear particularly justified during the most critical period of developmental plasticity when the child's immune, metabolic, and microbiological constitutions lend themselves to long-term adjustment. Pregnancy and early infancy epitomize an ideal developmental juncture for preventive measures aiming to reduce the risk of NCDs; by promoting the health of pregnant and lactating women today, the health of the next generation(s) may be successfully improved. The perfect tools for this initiative derive from the earliest and most massive source of environmental exposures, namely the microbiome and nutrition, due to their fundamental interactions in the function of the host immune and metabolic maturation.
Collapse
Affiliation(s)
- Erika Isolauri
- Department of Clinical Medicine, Faculty of Medicine, University of Turku, 20520 Turku, Finland;
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital, Kiinamyllynkatu 4-8, 20520 Turku, Finland
| | - Kirsi Laitinen
- Nutrition and Food Research Center & Institute of Biomedicine, Faculty of Medicine, University of Turku, 20520 Turku, Finland
| |
Collapse
|
46
|
Slater AS, Hickey RM, Davey GP. Interactions of human milk oligosaccharides with the immune system. Front Immunol 2025; 15:1523829. [PMID: 39877362 PMCID: PMC11772441 DOI: 10.3389/fimmu.2024.1523829] [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: 11/06/2024] [Accepted: 12/17/2024] [Indexed: 01/31/2025] Open
Abstract
Human milk oligosaccharides (HMOs) are abundant, diverse and complex sugars present in human breast milk. HMOs are well-characterized barriers to microbial infection and by modulating the human microbiome they are also thought to be nutritionally beneficial to the infant. The structural variety of over 200 HMOs, including neutral, fucosylated and sialylated forms, allows them to interact with the immune system in various ways. Clinically, HMOs impact allergic diseases, reducing autoimmune and inflammatory responses, and offer beneficial support to the preterm infant immune health. This review examines the HMO composition and associated immunomodulatory effects, including interactions with immune cell receptors and gut-associated immune responses. These immunomodulatory properties highlight the potential for HMO use in early stage immune development and for use as novel immunotherapeutics. HMO research is rapidly evolving and promises innovative treatments for immune-related conditions and improved health outcomes.
Collapse
Affiliation(s)
- Alanna S. Slater
- Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Rita M. Hickey
- Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
| | - Gavin P. Davey
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
47
|
Peng Z, Siziba LP, Mank M, Stahl B, Gonsalves J, Wernecke D, Rothenbacher D, Genuneit J. Profiles of 71 Human Milk Oligosaccharides and Novel Sub-Clusters of Type I Milk: Results from the Ulm SPATZ Health Study. Nutrients 2025; 17:280. [PMID: 39861410 PMCID: PMC11767774 DOI: 10.3390/nu17020280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/09/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Although approximately 160 human milk oligosaccharides (HMOs) have been identified, current studies on HMO quantitation are limited to the 10-19 most abundant HMOs. We assessed the variations in the relative concentrations of 71 HMO structures over lactation in human milk samples by an advanced liquid chromatography-mass spectrometry approach. METHODS Samples were collected from 64 mothers at 6 weeks, 6 months, and 12 months of lactation in the Ulm SPATZ Health Study, a German birth cohort. In this longitudinal study, we fitted linear mixed-effect models to analyze changes in the log2-transformed and standardized HMO concentration over time. Based on the profile of 71 HMOs, we also fitted a group-based multi-trajectory (GBMT) model to cluster mothers secreting cluster type I milk, who account for the majority of lactating mothers. RESULTS We found that 52 HMOs had a decreasing trend (regression coefficients ranging from -1.41 to -0.17) and 9 had an increasing trend (regression coefficients ranging from 0.25 to 0.64) during lactation, and the findings were statistically significant after multiple testing corrections. Using human milk samples of 49 mothers with type I milk, we further identified two novel sub-clusters with distinct longitudinal trajectories of concentrations of 71 HMOs during lactation: Type I-a (N = 20) and I-b (N = 29). These sub-clusters were not associated with maternal non-genetic characteristics. CONCLUSIONS Our findings extend existing knowledge about the structural diversity of HMOs and their variations over lactation. These may pave the way to investigate the potential nutritional benefits of various HMOs on infant health and early life development in the future.
Collapse
Affiliation(s)
- Zhuoxin Peng
- Pediatric Epidemiology, Department of Pediatrics, Medical Faculty, Leipzig University, Liebigstr 20a, Haus 6, 04103 Leipzig, Germany; (L.P.S.); (J.G.)
| | - Linda P. Siziba
- Pediatric Epidemiology, Department of Pediatrics, Medical Faculty, Leipzig University, Liebigstr 20a, Haus 6, 04103 Leipzig, Germany; (L.P.S.); (J.G.)
| | - Marko Mank
- Danone Research & Innovation, 3584 CT Utrecht, The Netherlands; (M.M.); (B.S.); (J.G.)
| | - Bernd Stahl
- Danone Research & Innovation, 3584 CT Utrecht, The Netherlands; (M.M.); (B.S.); (J.G.)
- Department of Chemical Biology & Drug Discovery, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - John Gonsalves
- Danone Research & Innovation, 3584 CT Utrecht, The Netherlands; (M.M.); (B.S.); (J.G.)
| | - Deborah Wernecke
- Institute of Epidemiology and Medical Biometry, Ulm University, 89075 Ulm, Germany; (D.W.); (D.R.)
- German Center for Child and Adolescent Health (DZKJ), 89075 Ulm, Germany
| | - Dietrich Rothenbacher
- Institute of Epidemiology and Medical Biometry, Ulm University, 89075 Ulm, Germany; (D.W.); (D.R.)
- German Center for Child and Adolescent Health (DZKJ), 89075 Ulm, Germany
| | - Jon Genuneit
- Pediatric Epidemiology, Department of Pediatrics, Medical Faculty, Leipzig University, Liebigstr 20a, Haus 6, 04103 Leipzig, Germany; (L.P.S.); (J.G.)
- Institute of Epidemiology and Medical Biometry, Ulm University, 89075 Ulm, Germany; (D.W.); (D.R.)
- German Center for Child and Adolescent Health (DZKJ), 04103 Leipzig, Germany
| |
Collapse
|
48
|
Weng WC, Liao HE, Chang CH, Hung SC, Du K, Tu Z, Lin CH, Ni CK. Unusual free trisaccharides in caprine colostrum discovered by logically derived sequence tandem mass spectrometry. Sci Rep 2025; 15:1586. [PMID: 39794344 PMCID: PMC11724002 DOI: 10.1038/s41598-024-81561-1] [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: 07/11/2024] [Accepted: 11/27/2024] [Indexed: 01/13/2025] Open
Abstract
Free oligosaccharides in human milk have many biological functions for infant health. The reducing end of most human milk oligosaccharides is lactose, and caprine milk was reported to contain oligosaccharides structurally similar to those present in human milk. The structures of oligosaccharides were traditionally determined using nuclear magnetic resonance spectroscopy or enzyme digestion followed by various detection methods, e.g., liquid. Mass spectrometry has much higher sensitivity than nuclear magnetic resonance spectroscopy and enzyme digestion. However, conventional mass spectrometry methods only determine part of the structures of oligosaccharides, i.e., compositions and linkage positions. In this study, we used the latest developed mass spectrometry method, namely logically derived sequence tandem mass spectrometry, to determine the complete structures (i.e., composition, linkage positions, anomericities, and stereoisomers) of free neutral trisaccharides in caprine colostrum and mature milk. The high sensitivity of mass spectrometry enables us to discover oligosaccharides of low abundance. Isomers of (Hex)2HexNAc, (Hex)3, and (Hex)2Fuc which have not been reported before were identified. Many of them do not have lactose at the reducing end. Instead, the reducing end is either Glcβ-(1-4)-Glc or Glcβ-(1-4)-GlcNAc. These unusual oligosaccharides are higher in concentration and more structurally diverse in caprine colostrum than that in caprine mature milk and human milk. The structural diversity indicates more complicated biosynthetic pathways of caprine milk compared to that of human milk.
Collapse
Affiliation(s)
- Wei-Chien Weng
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 106216, Taiwan
- Molecular Science and Technology, International Graduate Program, Department of Chemistry, Academia Sinica, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Hung-En Liao
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 106216, Taiwan
- Department of Appe of Figlied Chemistry, National Yang Ming Chiao Tung University, Hsinchu, 300093, Taiwan
| | - Cheng-Hsiu Chang
- Genomics Research Center, Academia Sinica, Taipei, 115201, Taiwan
| | - Shang-Cheng Hung
- Genomics Research Center, Academia Sinica, Taipei, 115201, Taiwan
| | - Kai Du
- Institute of Biological Chemistry, Academia Sinica, Taipei, 115201, Taiwan
| | - Zhijay Tu
- Institute of Biological Chemistry, Academia Sinica, Taipei, 115201, Taiwan
| | - Chun-Hung Lin
- Institute of Biological Chemistry, Academia Sinica, Taipei, 115201, Taiwan
- Department of Chemistry, Institute of Biochemical Sciences, National Taiwan University, Taipei, 106319, Taiwan
| | - Chi-Kung Ni
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 106216, Taiwan.
| |
Collapse
|
49
|
Huang C, Honda A, Suzuki T. Free oligosaccharides in serum. BBA ADVANCES 2025; 7:100139. [PMID: 39897077 PMCID: PMC11786756 DOI: 10.1016/j.bbadva.2025.100139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 02/04/2025] Open
Abstract
Glycans are sugars/sugar chains that are usually linked to proteins or lipids. The attachment of glycans often results in alterations of physicochemical/physiological properties of the carrier molecules, e.g., glycosylation of proteins can modulate their fate, intracellular localization, or interaction with cells/other proteins. On the other hand, unconjugated N-glycans (free N-glycans; FNGs) have been identified in the cytosol of eukaryotic cells. The processing pathway of intracellular FNGs has been clarified in recent years, but their biological functions remain unclear. Free oligosaccharides have also been identified in the sera of various animals. Structurally, these extracellular free glycans can be classified into three types: sialyl FNGs, oligomannose-type FNGs, and sialyl lactose/N-acetyllactosamine-type glycans. The extracellular FNGs show different structural features from intracellular FNGs, implying that their mechanism of formation is distinct. This mini-review summarizes current knowledge about the structures and formation mechanisms of free oligosaccharides in serum, and suggests their possible biological functions.
Collapse
Affiliation(s)
- Chengcheng Huang
- Chemical Glycobiology Laboratory, Institute for Glyco-core (iGOCRE), Tokai National Higher Education and Research System Nagoya University, Furo-cho, Nagoya, Aichi 464-8601, Japan
| | - Akinobu Honda
- Glycometabolic Biochemistry Laboratory, RIKEN-Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Tadashi Suzuki
- Glycometabolic Biochemistry Laboratory, RIKEN-Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| |
Collapse
|
50
|
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
|