1
|
Midtgaard SR, Hansen MS, Drachmann N, Geng X, Blans KIM, Møbjerg MMF, Frølund AF, Rasmussen JT, Ostenfeld MS. Industrial Scale Production and Characterization of a Whey Fraction Enriched in Extracellular Vesicle Material. JOURNAL OF EXTRACELLULAR BIOLOGY 2025; 4:e70044. [PMID: 40321664 PMCID: PMC12046291 DOI: 10.1002/jex2.70044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 02/25/2025] [Accepted: 03/01/2025] [Indexed: 05/08/2025]
Abstract
Human milk serves the sole nutritional role for the developing infant. During lactation, nano-sized extracellular vesicles (EVs) in milk containing a multitude of biologically active components are transferred from mother to offspring. Infant formula (IF) based on cow milk-derived ingredients has been reported to contain reduced levels of EVs as compared to human milk. There is therefore an unmet need to produce large-scale volumes of milk EVs to improve IF composition. Here, we report a scalable industrial production protocol for a bovine whey-derived ingredient that is highly enriched in EV material using a large-scale sequential ceramic membrane filtration setup. Furthermore, we demonstrate a robust and generally applicable analytical approach to determine the relative contributions of EVs and milk fat globule membrane (MFGM) using molar ratios of the membrane-bound proteins butyrophilin (BTN) and CD9 as surrogate markers for MFGM and EVs, respectively. Taken together, our findings provide a basis for comparing bovine milk-containing foods and aid in developing specialized ingredients that can minimize the compositional difference between infant formula and human milk.
Collapse
Affiliation(s)
| | | | | | - Xiaolu Geng
- Arla Foods Ingredients Group PSVibyCentral Jutland RegionDenmark
| | | | | | - Anny F. Frølund
- Arla Foods Ingredients Group PSVibyCentral Jutland RegionDenmark
| | | | | |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Geller F, Wu X, Lammi V, Abner E, Valliere JT, Nastou K, Rasmussen M, Andersson NW, Quinn L, DBDS Genomic Consortium, Aagaard B, Banasik K, Bliddal S, Boding L, Brunak S, Brøns N, Bybjerg-Grauholm J, Christoffersen LAN, Didriksen M, Dinh KM, Erikstrup C, Feldt-Rasmussen U, Grønbæk K, Kaspersen KA, Mikkelsen C, Nielsen CH, Nielsen HS, Nielsen SD, Nissen J, Sequeros CB, Tommerup N, Ullum H, Estonian Biobank Research Team, FinnGen, Spiliopoulos L, Bager P, Hviid A, Sørensen E, Pedersen OB, Lane JM, Lassaunière R, Ollila HM, Ostrowski SR, Feenstra B. Central role of glycosylation processes in human genetic susceptibility to SARS-CoV-2 infections with Omicron variants. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.21.24317689. [PMID: 39606378 PMCID: PMC11601703 DOI: 10.1101/2024.11.21.24317689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
The host genetics of SARS-CoV-2 has previously been studied based on cases from the earlier waves of the pandemic in 2020 and 2021, identifying 51 genomic loci associated with infection and/or severity. SARS-CoV-2 has shown rapid sequence evolution increasing transmissibility, particularly for Omicron variants, which raises the question whether this affected the host genetic factors. We performed a genome-wide association study of SARS-CoV-2 infection with Omicron variants including more than 150,000 cases from four cohorts. We identified 13 genome-wide significant loci, of which only five were previously described as associated with SARS-CoV-2 infection. The strongest signal was a single nucleotide polymorphism (SNP) intronic of ST6GAL1, a gene affecting immune development and function, and connected to three other associated loci (harboring MUC1, MUC5AC and MUC16) through O-glycan biosynthesis. We also found further evidence for an involvement of blood group systems in SARS-CoV-2 infection, as we observed association 1) for a different lead SNP in the ABO locus indicating a protective effect of blood group B against Omicron infection, 2) for the FUT2 SNP tagging secretor status also reported for SARS-CoV-2 infection with earlier variants, and 3) for the strongest expression quantitative trait locus (eQTL) for FUT3 (Lewis gene). Our study provides robust evidence for individual genetic variation related to glycosylation translating into susceptibility to SARS-CoV-2 infections with Omicron variants.
Collapse
Affiliation(s)
- Frank Geller
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Xiaoping Wu
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Vilma Lammi
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Erik Abner
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Jesse Tyler Valliere
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Molecular and Population Genetics Program, Broad Institute, Cambridge, MA, USA
| | - Katerina Nastou
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Morten Rasmussen
- Virus Research and Development Laboratory, Virus & Microbiological Special Diagnostics, Statens Serum Institut, Copenhagen, Denmark
| | | | - Liam Quinn
- Department of Clinical Immunology, Zealand University Hospital, Køge, Denmark
| | | | - Bitten Aagaard
- Department of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark
| | - Karina Banasik
- Department of Obstetrics and Gynecology, Copenhagen University Hospital, Amager & Hvidovre Hospital, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sofie Bliddal
- Department of Obstetrics and Gynecology, Copenhagen University Hospital, Amager & Hvidovre Hospital, Copenhagen, Denmark
- Department of Nephrology and Endocrinology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Lasse Boding
- Danish National Biobank, Statens Serum Institut, Copenhagen, Denmark
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nanna Brøns
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | | | - Lea Arregui Nordahl Christoffersen
- Department of Clinical Immunology, Zealand University Hospital, Køge, Denmark
- Institute of Biological Psychiatry, Mental Health Center Sct. Hans, Mental Health Services Copenhagen, Roskilde, Denmark
| | - Maria Didriksen
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Khoa Manh Dinh
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - Christian Erikstrup
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Ulla Feldt-Rasmussen
- Department of Nephrology and Endocrinology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Kirsten Grønbæk
- Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Christina Mikkelsen
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Novo Nordisk Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Claus Henrik Nielsen
- Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Henriette Svarre Nielsen
- Department of Obstetrics and Gynecology, Copenhagen University Hospital, Amager & Hvidovre Hospital, Copenhagen, Denmark
| | - Susanne Dam Nielsen
- Viro-Immunology Research Unit, Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Janna Nissen
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Celia Burgos Sequeros
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niels Tommerup
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | - Lampros Spiliopoulos
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Peter Bager
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Anders Hviid
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
- Pharmacovigilance Research Center, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Erik Sørensen
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Ole Birger Pedersen
- Department of Clinical Immunology, Zealand University Hospital, Køge, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jacqueline M Lane
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Molecular and Population Genetics Program, Broad Institute, Cambridge, MA, USA
| | - Ria Lassaunière
- Virus Research and Development Laboratory, Virus & Microbiological Special Diagnostics, Statens Serum Institut, Copenhagen, Denmark
| | - Hanna M Ollila
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Sisse Rye Ostrowski
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bjarke Feenstra
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
5
|
Panchal P, Rani R, Kumar R, Malik S, Mukesh M, Kaushik JK, Sodhi M, Mohanty AK, Kumar S. Optimizing workflow efficiency for analyzing low molecular weight endogenous peptides in colostrum. RSC Adv 2024; 14:29189-29200. [PMID: 39285878 PMCID: PMC11403347 DOI: 10.1039/d4ra03199g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 08/01/2024] [Indexed: 09/19/2024] Open
Abstract
Bovine milk and colostrum play pivotal roles in the nutritional support of both human and bovine infants. Colostrum, the initial milk secretion, is crucial for neonatal growth, providing essential nutrients, growth factors, immunity, and defense mechanisms through a diverse array of bioactive compounds, including bioactive proteins and peptides. Peptidomics, leveraging the potential health benefits of peptides derived from food and body fluids, has become prominent in contemporary research. Endogenous peptides (EPs) have gained notable scientific and commercial interest due to their potential biofunctional significance in areas such as immune health, antimicrobial, anti-inflammatory, antihypertensive, and antioxidative studies. In this investigation, we aimed to extract and analyze low molecular weight EPs from colostrum using four distinct peptide extraction methods, previously employed for EPs extraction from other bodily fluids. The efficiency of these methods was systematically compared and analysed to identify the most effective extraction technique for maximizing the identification of low molecular weight EPs from colostrum. This study represents a pioneering effort as no prior research has systematically compared different extraction methods for low molecular weight EPs from colostrum. Given the unique physical and chemical composition of colostrum compared to milk and other body fluids, a comprehensive analysis of EPs extraction methods was deemed essential. In the present study, we successfully extracted over 3200 EPs from colostrum using trichloroacetic acid (TCA) and a molecular weight cut off (MWCO) extraction method. The findings of this study revealed the extraction of EPs from colostrum, demonstrating potential inherent bioactivities as predicted by in silico tools.
Collapse
Affiliation(s)
- Priti Panchal
- ICAR-National Dairy Research Institute, Proteomics and Cell Biology Lab, Animal Biotechnology Division (ABTD) Karnal Haryana 132001 India
| | - Reeju Rani
- ICAR-National Dairy Research Institute, Proteomics and Cell Biology Lab, Animal Biotechnology Division (ABTD) Karnal Haryana 132001 India
| | - Rohit Kumar
- College of Public Health and Human Science, Oregon State University Corvalis OR 97331 USA
| | - Seema Malik
- ICAR-National Dairy Research Institute, Proteomics and Cell Biology Lab, Animal Biotechnology Division (ABTD) Karnal Haryana 132001 India
| | - Manishi Mukesh
- ICAR-National Bureau of Animal Genetic Resources Karnal Haryana 132001 India
| | - Jai Kumar Kaushik
- ICAR-National Dairy Research Institute, Proteomics and Cell Biology Lab, Animal Biotechnology Division (ABTD) Karnal Haryana 132001 India
| | - Monika Sodhi
- ICAR-National Bureau of Animal Genetic Resources Karnal Haryana 132001 India
| | | | - Sudarshan Kumar
- ICAR-National Dairy Research Institute, Proteomics and Cell Biology Lab, Animal Biotechnology Division (ABTD) Karnal Haryana 132001 India
| |
Collapse
|
6
|
Pados BF, Camp L. Physiology of Human Lactation and Strategies to Support Milk Supply for Breastfeeding. Nurs Womens Health 2024; 28:303-314. [PMID: 38972331 DOI: 10.1016/j.nwh.2024.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 05/21/2024] [Indexed: 07/09/2024]
Abstract
Despite advances across the globe in breastfeeding initiation rates, many families continue to report they are not meeting their breastfeeding goals. Concerns about milk supply, infant nutritional intake, and infant weight gain are among the most commonly cited reasons for early breastfeeding cessation. Nurses working with individuals during the perinatal period are uniquely positioned to educate families and offer evidence-based interventions to promote optimal milk supply, infant growth, and maternal mental and physical health. Such interventions include early and frequent skin-to-skin care, emptying of the breast, and professional lactation support. By implementing such evidence-based practices in the first hours after birth and connecting families to lactation support in the first 14 days, nurses can begin to help families achieve their breastfeeding goals.
Collapse
|
7
|
Han M, Li K, Fang Y, Chen B, Sun H, Xie Q, Zhang S, Jiang S, Lv J, Xu Y. Characteristics of casein phosphopeptides in Chinese human milk and its correlation with infant growth: A cross-sectional study. Food Chem 2024; 447:139007. [PMID: 38518618 DOI: 10.1016/j.foodchem.2024.139007] [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: 11/16/2023] [Revised: 03/04/2024] [Accepted: 03/09/2024] [Indexed: 03/24/2024]
Abstract
This research aimed to investigate the characteristics of casein phosphopeptides in Chinese human milk, and their potential relationship to infant growth. Using the liquid chromatography-Orbitrap-mass spectrometry technique, a total of 15 casein phosphopeptides were identified from 200 human milk samples. Also, our results indicate that casein phosphopeptides were phosphorylated with only one phosphate. The relative concentrations of casein phosphopeptides at 6 months postpartum were increased compared with milk at 2 months (FDR < 0.05). Significantly positive correlations were observed between casein phosphopeptides and infant growth, as shown by four casein phosphopeptides were positively correlated with the infants' weight-for-age Z-scores (rs range from 0.20 to 0.29), and three casein phosphopeptides were positively correlated with the infants' length-for-age Z-scores (rs range from 0.19 to 0.27). This study is the first to reveal the phosphorylated level and composition of casein phosphopeptides in Chinese human milk, and their potential relationship with infant growth.
Collapse
Affiliation(s)
- Muke Han
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100083, China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100083, China
| | - Kaifeng Li
- Feihe Research Institute, Heilongjiang Feihe Dairy Co., Ltd, Beijing 100016, China; PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, Beijing 100083, China
| | - Yuehui Fang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Baorong Chen
- Institute of Food Science and Technology, Chinese Academy of Agricultural Science, Beijing 100081, China
| | - Han Sun
- Feihe Research Institute, Heilongjiang Feihe Dairy Co., Ltd, Beijing 100016, China; PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, Beijing 100083, China
| | - Qinggang Xie
- Feihe Research Institute, Heilongjiang Feihe Dairy Co., Ltd, Beijing 100016, China; PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, Beijing 100083, China
| | - Shuwen Zhang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Science, Beijing 100081, China
| | - Shilong Jiang
- Feihe Research Institute, Heilongjiang Feihe Dairy Co., Ltd, Beijing 100016, China; PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, Beijing 100083, China.
| | - Jiaping Lv
- Institute of Food Science and Technology, Chinese Academy of Agricultural Science, Beijing 100081, China.
| | - Yajun Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100083, China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100083, China.
| |
Collapse
|
8
|
Ning J, Yang M, Zhu Q, Liu X, Li M, Luo X, Yue X. Revealing the diversity of endogenous peptides and parent proteins in human colostrum and mature milk through peptidomics analysis. Food Chem 2024; 445:138651. [PMID: 38359565 DOI: 10.1016/j.foodchem.2024.138651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/17/2024]
Abstract
Endogenous peptides and their parent proteins are important nutritional components with diverse biological functions. The objective of this study was to analyze and compare endogenous peptides and parent proteins found in human colostrum (HC) and human mature milk (HM) using a 4D label-free technique. In total, 5162 and 940 endogenous peptides derived from 258 parent proteins were identified in human milk by database (DB) search and de novo, respectively. Among these peptides, 2446 differentially expressed endogenous peptides with various bioactivities were identified. The Gene Ontology analysis unveiled the cellular components, biological processes, and molecular functions associated with these parent proteins. Metabolic pathway analysis suggested that neutrophil extracellular trap formation had the greatest significance with 24 parent proteins. These findings will offer a fresh perspective on the development of infant formula powder, highlighting the potential for incorporating these changes to enhance its nutritional composition and benefits.
Collapse
Affiliation(s)
- Jianting Ning
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Mei Yang
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Qing Zhu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Xiaoyu Liu
- Department of Obstetrics and Gynaecology, General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Mohan Li
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Xue Luo
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China.
| | - Xiqing Yue
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China.
| |
Collapse
|
9
|
Thesbjerg MN, Sundekilde UK, Poulsen NA, Larsen LB, Nielsen SDH. A novel proteomic approach for the identification and relative quantification of disulfide-bridges in the human milk proteome. J Proteomics 2024; 301:105194. [PMID: 38723850 DOI: 10.1016/j.jprot.2024.105194] [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: 11/29/2023] [Revised: 04/26/2024] [Accepted: 05/05/2024] [Indexed: 05/13/2024]
Abstract
This study explores the disulfide bridges present in the human milk proteome by a novel approach permitting both positional identification and relative quantification of the disulfide bridges. Human milk from six donors was subjected to trypsin digestion without reduction. The digested human milk proteins were analyzed by nanoLC-timsTOF Pro combined with data analysis using xiSEARCH. A total of 85 unique disulfide bridges were identified in 25 different human milk proteins. The total relative abundance of disulfide bridge-containing peptides constituted approximately 5% of the total amount of tryptic-peptides. Seven inter-molecular disulfide bridges were identified between either α-lactalbumin and lactotransferrin (5) or αS1-casein and κ-casein (2). All cysteines involved in the observed disulfide bridges of α-lactalbumin and lactotransferrin were mapped onto protein models using AlphaFold2 Multimer to estimate the length of the observed disulfide bridges. The lengths of the disulfide bridges of lactotransferrin indicate a potential for multi- or poly-merization of lactotransferrin. The high number of intramolecular lactotransferrin disulfide bridges identified, suggests that these are more heterogeneous than previously presumed. SIGNIFICANCE: Disulfide-bridges in the human milk proteome are an often overseen post-transaltional modification. Thus, mapping the disulfide-bridges, their positions and relative abundance, are valuable new knowledge needed for an improved understanding of human milk protein behaviour. Although glycosylation and phosphorylation have been described, even less information is available on the disulfide bridges and the disulfide-bridge derived protein complexes. This is important for future work in precision fermentation for recombinant production of human milk proteins, as this will highlight which disulfide-bridges are naturally occouring in human milk proteins. Further, this knowledge would be of value for the infant formula industry as it provides more information on how to humanize bovine-milk based infant formula. The novel method developed here can be broadly applied in other biological systems as the disulfid-brigdes are important for the structure and functionality of proteins.
Collapse
Affiliation(s)
- Martin Nørmark Thesbjerg
- Department of Food Science, Aarhus University, Agro Food Park 48, DK-8200 Aarhus N, Denmark; Sino-Danish College (SDC), University of Chinese Academy of Science, Huairou District, Beijing 101408, China.
| | | | - Nina Aagaard Poulsen
- Department of Food Science, Aarhus University, Agro Food Park 48, DK-8200 Aarhus N, Denmark
| | - Lotte Bach Larsen
- Department of Food Science, Aarhus University, Agro Food Park 48, DK-8200 Aarhus N, Denmark
| | | |
Collapse
|
10
|
Sabino J, Tarassishin L, Eisele C, Hawkins K, Barré A, Nair N, Rendon A, Debebe A, Picker M, Agrawal M, Stone J, George J, Legnani P, Maser E, Chen CL, Thjømøe A, Mørk E, Dubinsky M, Hu J, Colombel JF, Peter I, Torres J. Influence of Early Life Factors, including breast milk Composition, on the Microbiome of Infants Born to Mothers with and without Inflammatory Bowel Disease. J Crohns Colitis 2023; 17:1723-1732. [PMID: 37279927 PMCID: PMC10673817 DOI: 10.1093/ecco-jcc/jjad096] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/29/2023] [Accepted: 06/05/2023] [Indexed: 06/08/2023]
Abstract
BACKGROUND AND AIMS Herein we analysed the influence of early life factors, including breast milk composition, on the development of the intestinal microbiota of infants born to mothers with and without IBD. METHODS The MECONIUM [Exploring MEChanisms Of disease traNsmission In Utero through the Microbiome] study is a prospective cohort study consisting of pregnant women with or without IBD and their infants. Longitudinal stool samples were collected from babies and analysed using 16s rRNA sequencing and faecal calprotectin. Breast milk proteomics was profiled using Olink inflammation panel. RESULTS We analysed gut microbiota of 1034 faecal samples from 294 infants [80 born to mothers with and 214 to mothers without IBD]. Alpha diversity was driven by maternal IBD status and time point. The major influencers of the overall composition of the microbiota were mode of delivery, feeding, and maternal IBD status. Specific taxa were associated with these exposures, and maternal IBD was associated with a reduction in Bifidobacterium. In 312 breast milk samples [91 from mothers with IBD], mothers with IBD displayed lower abundance of proteins involved in immune regulation, such as thymic stromal lymphopoietin, interleukin-12 subunit beta, tumour necrosis factor-beta, and C-C motif chemokine 20, as compared with control mothers [adjusted p = 0.0016, 0.049, 0.049, and 0.049, respectively], with negative correlations with baby´s calprotectin, and microbiome at different time points. CONCLUSION Maternal IBD diagnosis influences microbiota in their offspring during early life. The proteomic profile of breast milk of women with IBD differs from that of women without IBD, with distinct time-dependent associations with baby's gut microbiome and feacal calprotectin.
Collapse
Affiliation(s)
- João Sabino
- Icahn School of Medicine at Mount Sinai, Department of Genetics and Genomic Sciences, New York, NY, USA
- Icahn School of Medicine at Mount Sinai, Division of Gastroenterology, Department of Medicine, New York, NY, USA
- Department of Gastroenterology, University Hospitals of Leuven, Leuven, Belgium
| | - Leonid Tarassishin
- Icahn School of Medicine at Mount Sinai, Department of Genetics and Genomic Sciences, New York, NY, USA
| | - Caroline Eisele
- Icahn School of Medicine at Mount Sinai, Department of Genetics and Genomic Sciences, New York, NY, USA
- College of Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Kelly Hawkins
- Icahn School of Medicine at Mount Sinai, Department of Genetics and Genomic Sciences, New York, NY, USA
| | - Amelie Barré
- Icahn School of Medicine at Mount Sinai, Department of Genetics and Genomic Sciences, New York, NY, USA
| | - Nile Nair
- Icahn School of Medicine at Mount Sinai, Department of Genetics and Genomic Sciences, New York, NY, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Alexa Rendon
- Icahn School of Medicine at Mount Sinai, Department of Genetics and Genomic Sciences, New York, NY, USA
| | - Anketse Debebe
- Icahn School of Medicine at Mount Sinai, Department of Genetics and Genomic Sciences, New York, NY, USA
| | - Mellissa Picker
- Icahn School of Medicine at Mount Sinai, Department of Genetics and Genomic Sciences, New York, NY, USA
| | - Manasi Agrawal
- Icahn School of Medicine at Mount Sinai, Division of Gastroenterology, Department of Medicine, New York, NY, USA
- Center for Molecular Prediction of IBD [PREDICT], Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Joanne Stone
- Gastroenterology Division, Icahn School of Medicine at Mount Sinai, Department of Obstetrics, Gynecology and Reproductive Sciences, New York, NY, USA
| | - James George
- Icahn School of Medicine at Mount Sinai, Division of Gastroenterology, Department of Medicine, New York, NY, USA
| | - Peter Legnani
- Icahn School of Medicine at Mount Sinai, Division of Gastroenterology, Department of Medicine, New York, NY, USA
| | - Elana Maser
- Icahn School of Medicine at Mount Sinai, Division of Gastroenterology, Department of Medicine, New York, NY, USA
| | - Ching-Lynn Chen
- Gastroenterology Division, Icahn School of Medicine at Mount Sinai, Department of Obstetrics, Gynecology and Reproductive Sciences, New York, NY, USA
| | | | | | - Marla Dubinsky
- Icahn School of Medicine at Mount Sinai, Division of Pediatric Gastroenterology and Hepatology, New York, NY, USA
| | - Jianzhong Hu
- Icahn School of Medicine at Mount Sinai, Department of Genetics and Genomic Sciences, New York, NY, USA
| | - Jean-Frederic Colombel
- Icahn School of Medicine at Mount Sinai, Division of Gastroenterology, Department of Medicine, New York, NY, USA
| | - Inga Peter
- Icahn School of Medicine at Mount Sinai, Department of Genetics and Genomic Sciences, New York, NY, USA
| | - Joanna Torres
- Icahn School of Medicine at Mount Sinai, Division of Gastroenterology, Department of Medicine, New York, NY, USA
- Gastroenterology Division, Hospital Beatriz Ângelo, Loures, Portugal
- Gastroenterology Division, Hospital da Luz, Lisbon, Portugal
- Faculdade de Medicina, Universidade de Lisboa, Portugal
| |
Collapse
|
11
|
Thesbjerg MN, Nielsen SDH, Sundekilde UK, Poulsen NA, Larsen LB. Fingerprinting of Proteases, Protease Inhibitors and Indigenous Peptides in Human Milk. Nutrients 2023; 15:4169. [PMID: 37836453 PMCID: PMC10574734 DOI: 10.3390/nu15194169] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/19/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
The presence of proteases and their resulting level of activity on human milk (HM) proteins may aid in the generation of indigenous peptides as part of a pre-digestion process, of which some have potential bioactivity for the infant. The present study investigated the relative abundance of indigenous peptides and their cleavage products in relation to the abundance of observed proteases and protease inhibitors. The proteomes and peptidomes in twelve HM samples, representing six donors at lactation months 1 and 3, were profiled. In the proteome, 39 proteases and 29 protease inhibitors were identified in 2/3 of the samples. Cathepsin D was found to be present in higher abundance in the proteome compared with plasmin, while peptides originating from plasmin cleavage were more abundant than peptides from cathepsin D cleavage. As both proteases are present as a system of pro- and active- forms, their activation indexes were calculated. Plasmin was more active in lactation month 3 than month 1, which correlated with the total relative abundance of the cleavage product ascribed to plasmin. By searching the identified indigenous peptides in the milk bioactive peptide database, 283 peptides were ascribed to 10 groups of bioactivities. Antimicrobial peptides were significantly more abundant in month 1 than month 3; this group comprised 103 peptides, originating from the β-CN C-terminal region.
Collapse
Affiliation(s)
- Martin Nørmark Thesbjerg
- Department of Food Science, Aarhus University, Agro Food Park 48, DK-8200 Aarhus, Denmark; (M.N.T.); (N.A.P.)
- Sino-Danish College (SDC), University of Chinese Academy of Sciences, Huairou District, Beijing 101408, China
| | - Søren Drud-Heydary Nielsen
- Department of Food Science, Aarhus University, Agro Food Park 48, DK-8200 Aarhus, Denmark; (M.N.T.); (N.A.P.)
| | - Ulrik Kræmer Sundekilde
- Department of Food Science, Aarhus University, Agro Food Park 48, DK-8200 Aarhus, Denmark; (M.N.T.); (N.A.P.)
| | - Nina Aagaard Poulsen
- Department of Food Science, Aarhus University, Agro Food Park 48, DK-8200 Aarhus, Denmark; (M.N.T.); (N.A.P.)
| | - Lotte Bach Larsen
- Department of Food Science, Aarhus University, Agro Food Park 48, DK-8200 Aarhus, Denmark; (M.N.T.); (N.A.P.)
| |
Collapse
|
12
|
Analysis of milk with liquid chromatography–mass spectrometry: a review. Eur Food Res Technol 2023. [DOI: 10.1007/s00217-022-04197-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
AbstractAs a widely consumed foodstuff, milk and dairy products are increasingly studied over the years. At the present time, milk profiling is used as a benchmark to assess the properties of milk. Modern biomolecular mass spectrometers have become invaluable to fully characterize the milk composition. This review reports the analysis of milk and its components using liquid chromatography coupled with mass spectrometry (LC–MS). LC–MS analysis as a whole will be discussed subdivided into the major constituents of milk, namely, lipids, proteins, sugars and the mineral fraction.
Collapse
|
13
|
Dingess KA, Hoek M, van Rijswijk DMH, Tamara S, den Boer MA, Veth T, Damen MJA, Barendregt A, Romijn M, Juncker HG, van Keulen BJ, Vidarsson G, van Goudoever JB, Bondt A, Heck AJR. Identification of common and distinct origins of human serum and breastmilk IgA1 by mass spectrometry-based clonal profiling. Cell Mol Immunol 2023; 20:26-37. [PMID: 36447030 PMCID: PMC9707141 DOI: 10.1038/s41423-022-00954-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 11/03/2022] [Indexed: 11/30/2022] Open
Abstract
The most abundant immunoglobulin present in the human body is IgA. It has the highest concentrations at the mucosal lining and in biofluids such as milk and is the second most abundant class of antibodies in serum. We assessed the structural diversity and clonal repertoire of IgA1-containing molecular assemblies longitudinally in human serum and milk from three donors using a mass spectrometry-based approach. IgA-containing molecules purified from serum or milk were assessed by the release and subsequent analysis of their Fab fragments. Our data revealed that serum IgA1 consists of two distinct structural populations, namely monomeric IgA1 (∼80%) and dimeric joining (J-) chain coupled IgA1 (∼20%). Also, we confirmed that IgA1 in milk is present solely as secretory (S)IgA, consisting of two (∼50%), three (∼33%) or four (∼17%) IgA1 molecules assembled with a J-chain and secretory component (SC). Interestingly, the serum and milk IgA1-Fab repertoires were distinct between monomeric, and J-chain coupled dimeric IgA1. The serum dimeric J-chain coupled IgA1 repertoire contained several abundant clones also observed in the milk IgA1 repertoire. The latter repertoire had little to no overlap with the serum monomeric IgA1 repertoire. This suggests that human IgA1s have (at least) two distinct origins; one of these produces dimeric J-chain coupled IgA1 molecules, shared in human serum and milk, and another produces monomeric IgA1 ending up exclusively in serum.
Collapse
Affiliation(s)
- Kelly A Dingess
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, Utrecht, 3584 CH, The Netherlands
- Netherlands Proteomics Center, Padualaan 8, Utrecht, 3584 CH, The Netherlands
- Amsterdam UMC, Vrije Universiteit, University of Amsterdam, Emma Children's Hospital, Amsterdam Reproduction & Development Research Institute, Department of Pediatrics, Amsterdam, the Netherlands
| | - Max Hoek
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, Utrecht, 3584 CH, The Netherlands
- Netherlands Proteomics Center, Padualaan 8, Utrecht, 3584 CH, The Netherlands
| | - Danique M H van Rijswijk
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, Utrecht, 3584 CH, The Netherlands
- Netherlands Proteomics Center, Padualaan 8, Utrecht, 3584 CH, The Netherlands
| | - Sem Tamara
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, Utrecht, 3584 CH, The Netherlands
- Netherlands Proteomics Center, Padualaan 8, Utrecht, 3584 CH, The Netherlands
| | - Maurits A den Boer
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, Utrecht, 3584 CH, The Netherlands
- Netherlands Proteomics Center, Padualaan 8, Utrecht, 3584 CH, The Netherlands
| | - Tim Veth
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, Utrecht, 3584 CH, The Netherlands
- Netherlands Proteomics Center, Padualaan 8, Utrecht, 3584 CH, The Netherlands
| | - Mirjam J A Damen
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, Utrecht, 3584 CH, The Netherlands
- Netherlands Proteomics Center, Padualaan 8, Utrecht, 3584 CH, The Netherlands
| | - Arjan Barendregt
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, Utrecht, 3584 CH, The Netherlands
- Netherlands Proteomics Center, Padualaan 8, Utrecht, 3584 CH, The Netherlands
| | - Michelle Romijn
- Amsterdam UMC, Vrije Universiteit, University of Amsterdam, Emma Children's Hospital, Amsterdam Reproduction & Development Research Institute, Department of Pediatrics, Amsterdam, the Netherlands
| | - Hannah G Juncker
- Amsterdam UMC, Vrije Universiteit, University of Amsterdam, Emma Children's Hospital, Amsterdam Reproduction & Development Research Institute, Department of Pediatrics, Amsterdam, the Netherlands
| | - Britt J van Keulen
- Amsterdam UMC, Vrije Universiteit, University of Amsterdam, Emma Children's Hospital, Amsterdam Reproduction & Development Research Institute, Department of Pediatrics, Amsterdam, the Netherlands
| | - Gestur Vidarsson
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Johannes B van Goudoever
- Amsterdam UMC, Vrije Universiteit, University of Amsterdam, Emma Children's Hospital, Amsterdam Reproduction & Development Research Institute, Department of Pediatrics, Amsterdam, the Netherlands
| | - Albert Bondt
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, Utrecht, 3584 CH, The Netherlands
- Netherlands Proteomics Center, Padualaan 8, Utrecht, 3584 CH, The Netherlands
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, Utrecht, 3584 CH, The Netherlands.
- Netherlands Proteomics Center, Padualaan 8, Utrecht, 3584 CH, The Netherlands.
| |
Collapse
|
14
|
Moya-Alvarez V, Eussen SRBM, Mank M, Koyembi JCJ, Nyasenu YT, Ngaya G, Mad-Bondo D, Kongoma JB, Stahl B, Sansonetti PJ, Bourdet-Sicard R. Human milk nutritional composition across lactational stages in Central Africa. Front Nutr 2022; 9:1033005. [PMID: 36466422 PMCID: PMC9709887 DOI: 10.3389/fnut.2022.1033005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/27/2022] [Indexed: 03/11/2024] Open
Abstract
The African region encompasses the highest undernutrition burden with the highest neonatal and infant mortality rates globally. Under these circumstances, breastfeeding is one of the most effective ways to ensure child health and development. However, evidence on human milk (HM) composition from African women is scarce. This is of special concern, as we have no reference data from HM composition in the context of food insecurity in Africa. Furthermore, data on the evolution of HM across lactational stages in this setting lack as well. In the MITICA study, we conducted a cohort study among 48 Central-African women and their 50 infants to analyze the emergence of gut dysbiosis in infants and describe the mother-infant transmission of microbiota between birth and 6 months of age. In this context, we assessed nutritional components in HM of 48 lactating women in Central Africa through five sampling times from week 1 after birth until week 25. Unexpectedly, HM-type III (Secretor + and Lewis genes -) was predominant in HM from Central African women, and some nutrients differed significantly among HM-types. While lactose concentration increased across lactation periods, fatty acid concentration did not vary significantly. The overall median level of 16 detected individual human milk oligosaccharides (HMOs; core structures as well as fucosylated and sialylated ones) decreased from 7.3 g/l at week 1 to 3.5 g/l at week 25. The median levels of total amino acids in HM dropped from 12.8 mg/ml at week 1 to 7.4 mg/ml at week 25. In contrast, specific free amino acids increased between months 1 and 3 of lactation, e.g., free glutamic acid, glutamine, aspartic acid, and serine. In conclusion, HM-type distribution and certain nutrients differed from Western mother HM.
Collapse
Affiliation(s)
- Violeta Moya-Alvarez
- Unité de Pathogénie Microbienne Moléculaire, INSERM U1202, Department of Cell Biology and Infection, Institut Pasteur, Paris, France
- Epidemiology of Emergent Diseases Unit, Global Health Department, Institut Pasteur, Paris, France
| | - Simone R. B. M. Eussen
- Human Milk Research and Analytical Science, Danone Nutricia Research, Utrecht, Netherlands
| | - Marko Mank
- Human Milk Research and Analytical Science, Danone Nutricia Research, Utrecht, Netherlands
| | | | - Yawo Tufa Nyasenu
- Laboratoire d'Analyses Médicales, Institut Pasteur de Bangui, Bangui, Central African Republic
- Laboratoire de Biologie Moléculaire et d'Immunologie, Université de Lomé, Lomé, Togo
| | - Gilles Ngaya
- Laboratoire de Biologie Moléculaire et d'Immunologie, Université de Lomé, Lomé, Togo
| | - Daniel Mad-Bondo
- Direction du Service de Santé de la Gendarmerie, Sis Camp Henri Izamo, Bangui, Central African Republic
| | - Jean-Bertrand Kongoma
- Direction du Service de Santé de la Gendarmerie, Sis Camp Henri Izamo, Bangui, Central African Republic
| | - Bernd Stahl
- Human Milk Research and Analytical Science, Danone Nutricia Research, Utrecht, Netherlands
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Philippe J. Sansonetti
- Unité de Pathogénie Microbienne Moléculaire, INSERM U1202, Department of Cell Biology and Infection, Institut Pasteur, Paris, France
- Chaire de Microbiologie et Maladies Infectieuses, Collège de France, Paris, France
| | | |
Collapse
|
15
|
Peptidomics as a tool to analyze endogenous peptides in milk and milk-related peptides. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2022.102199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
|
16
|
Sun X, Yu Z, Liang C, Xie S, Wang H, Wang J, Yang Y, Han R. Comparative analysis of changes in whey proteins of goat milk throughout the lactation cycle using quantitative proteomics. J Dairy Sci 2022; 106:792-806. [DOI: 10.3168/jds.2022-21800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 08/25/2022] [Indexed: 11/23/2022]
|
17
|
Remoroza CA, Burke MC, Yang X, Sheetlin S, Mirokhin Y, Markey SP, Tchekhovskoi DV, Stein SE. Mass Spectral Library Methods for Analysis of Site-Specific N-Glycosylation: Application to Human Milk Proteins. J Proteome Res 2022; 21:2421-2434. [DOI: 10.1021/acs.jproteome.2c00286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Concepcion A. Remoroza
- Mass Spectrometry Data Center, Biomolecular Measurement Division, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, Maryland 20899, United States
| | - Meghan C. Burke
- Mass Spectrometry Data Center, Biomolecular Measurement Division, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, Maryland 20899, United States
| | - Xiaoyu Yang
- Mass Spectrometry Data Center, Biomolecular Measurement Division, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, Maryland 20899, United States
| | - Sergey Sheetlin
- Mass Spectrometry Data Center, Biomolecular Measurement Division, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, Maryland 20899, United States
| | - Yuri Mirokhin
- Mass Spectrometry Data Center, Biomolecular Measurement Division, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, Maryland 20899, United States
| | - Sanford P. Markey
- Mass Spectrometry Data Center, Biomolecular Measurement Division, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, Maryland 20899, United States
| | - Dmitrii V. Tchekhovskoi
- Mass Spectrometry Data Center, Biomolecular Measurement Division, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, Maryland 20899, United States
| | - Stephen E. Stein
- Mass Spectrometry Data Center, Biomolecular Measurement Division, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, Maryland 20899, United States
| |
Collapse
|
18
|
Zheng Y, Correa-Silva S, Palmeira P, Carneiro-Sampaio M. Maternal vaccination as an additional approach to improve the protection of the nursling: Anti-infective properties of breast milk. Clinics (Sao Paulo) 2022; 77:100093. [PMID: 35963149 PMCID: PMC9382412 DOI: 10.1016/j.clinsp.2022.100093] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/08/2022] [Accepted: 07/14/2022] [Indexed: 11/30/2022] Open
Abstract
Human milk constitutes a secretion with unique functions of both nourishing the nursling and providing protection against enteric and respiratory infections, mainly due to its content of secretory IgA antibodies but also due to the presence of a plethora of bioactive factors. Specific IgA antibodies are produced locally by plasma cells derived from B lymphocytes that migrate from other mucosae to the mammary gland during lactation, particularly from the gastrointestinal and respiratory tracts. Therefore, here, the authors will provide a comprehensive review of the content and functions of different nutritional and bioactive anti-infectious components from breast milk, such as oligosaccharides, lactoferrin, haptocorrin, α-lactalbumin, k-casein, lysozyme, lactoperoxidase, mucin, fatty acids, defensins, cytokines and chemokines, hormones and growth factors, complement proteins, leukocytes and nucleic acids, including microRNAs, among many others, and the induction of antibody responses in breast milk after maternal vaccination with several licensed vaccines, including the anti-SARS-CoV-2 vaccine preparations used worldwide. Currently, in the midst of the pandemic, maternal vaccination has re-emerged as a crucial source of passive immunity to the neonate through the placenta and breastfeeding, considering that maternal vaccination can induce specific antibodies if performed during pregnancy and after delivery. There have been some reports in the literature about milk IgA antibodies induced by bacterial antigens or inactivated virus vaccines, such as anti-diphtheria-tetanus-pertussis, anti-influenza viruses, anti-pneumococcal and meningococcal polysaccharide preparations. Regarding anti-SARS-CoV-2 vaccines, most studies demonstrate elevated levels of specific IgA and IgG antibodies in milk with virus-neutralizing ability after maternal vaccination, which represents an additional approach to improve the protection of the nursling during the entire breastfeeding period.
Collapse
Affiliation(s)
- Yingying Zheng
- Department of Pediatrics, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil.
| | - Simone Correa-Silva
- Department of Pediatrics, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil; Universidade Paulista, UNIP, São Paulo, SP, Brazil.
| | - Patricia Palmeira
- Laboratory of Medical Investigation (LIM-36), Department of Pediatrics, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil.
| | - Magda Carneiro-Sampaio
- Department of Pediatrics, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
19
|
Zhu J, Yu X, Wang Y, Bai S, Lai J, Tong X, Xing Y. Longitudinal changes of lactopontin (milk osteopontin) in term and preterm human milk. Front Nutr 2022; 9:962802. [PMID: 35967792 PMCID: PMC9372532 DOI: 10.3389/fnut.2022.962802] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/13/2022] [Indexed: 11/24/2022] Open
Abstract
Background Lactopontin (LPN) in breast milk, also known as milk osteopontin is thought to play a myriad of important roles in infants when they are immature. The purpose of the present study was to examine the longitudinal changes in LPN concentrations in term and preterm milk, and elucidate the links between maternal characteristics, LPN levels, and child growth in a birth cohort. Methods 131 mothers who delivered term, moderate-late preterm (MPT), very preterm (VPT), and extremely preterm (EPT) infants were included, milk samples were collected at 7, 14, 28, and 120 days postpartum. LPN concentration was determined by multiple reaction monitoring (MRM) using ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS). Results Our results indicated that LPN change over time of VPT (P = 0.024) and EPT (P = 0.003) were significantly different from term milk, although they all gradually decreased with lactation. In terms of LPN-related factors, maternal age was a significant contributor in late mature milk and pre-pregnancy BMI a significant contributor to colostrum and transitional milk. We further investigated relationships between LPN levels and infant weight and our results suggested that high levels of LPN in breast milk might be useful for the catch-up growth of infants. Conclusion LPN levels in breast milk are related to maternal factors, and differences in LPN levels may affect the growth of infants. As milk is a critical part in the mother-breastmilk-infant "triad," the association between maternal-infant factors and milk LPN levels warrants further study.
Collapse
Affiliation(s)
- Jing Zhu
- Beijing Institute of Nutritional Resources, Beijing Academy of Science and Technology, Beijing, China
| | - Xue Yu
- School of Public Health, Peking University Health Science Center, Beijing, China
| | - Yiran Wang
- Beijing Institute of Nutritional Resources, Beijing Academy of Science and Technology, Beijing, China
| | - Shasha Bai
- Beijing Institute of Nutritional Resources, Beijing Academy of Science and Technology, Beijing, China
| | - Jianqiang Lai
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiaomei Tong
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| | - Yan Xing
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| |
Collapse
|
20
|
Zhu J, Liu M, Xing Y. Preterm birth and human milk proteome: are we ready for individualized fortification? Curr Opin Clin Nutr Metab Care 2022; 25:216-222. [PMID: 35199659 DOI: 10.1097/mco.0000000000000824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW Preterm birth is one of the most pressing clinical problems in obstetrics and neonatology worldwide. One of the most sophisticated components of human milk is the proteome and a better understanding of it can lead to precision guides for feeding preterm infants. In this review, we will examine recent research focused on the human milk proteome and individualized protein fortification of human milk. RECENT FINDINGS In both preterm and term birth, the protein content in mother's own milk dropped rapidly in the early postnatal period. Preterm milk had a higher protein content and contained different protein and endogenous peptide compositions compared with term milk. The peptides in gastrointestinal fluids of preterm infants still need further investigation. Individualized fortification is more superior to standard fortification, but it only focuses on the total protein amount. There is no data concerning the composition and posttranslational modifications of proteins and endogenous peptides with fortification and their longterm effects. SUMMARY Comprehensive identification and characterization of the human milk proteome have led to the targeted breast milk fortification theory of preterm infant feeding and has also been enriched by clinical trials. However, to achieve fortification of key proteins and/or endogenous peptides, as standard clinical practice, requires additional studies. Future research should explore the long-term effect of protein fortification and pay more attention to quality rather than quantity in relation to infant body composition and growth outcomes.
Collapse
Affiliation(s)
- Jing Zhu
- Beijing Institute of Nutritional Resources, Beijing Academy of Science and Technology
| | - Mengyuan Liu
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| | - Yan Xing
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| |
Collapse
|
21
|
Nyquist SK, Gao P, Haining TKJ, Retchin MR, Golan Y, Drake RS, Kolb K, Mead BE, Ahituv N, Martinez ME, Shalek AK, Berger B, Goods BA. Cellular and transcriptional diversity over the course of human lactation. Proc Natl Acad Sci U S A 2022; 119:e2121720119. [PMID: 35377806 PMCID: PMC9169737 DOI: 10.1073/pnas.2121720119] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/14/2022] [Indexed: 12/04/2022] Open
Abstract
Human breast milk (hBM) is a dynamic fluid that contains millions of cells, but their identities and phenotypic properties are poorly understood. We generated and analyzed single-cell RNA-sequencing (scRNA-seq) data to characterize the transcriptomes of cells from hBM across lactational time from 3 to 632 d postpartum in 15 donors. We found that the majority of cells in hBM are lactocytes, a specialized epithelial subset, and that cell-type frequencies shift over the course of lactation, yielding greater epithelial diversity at later points. Analysis of lactocytes reveals a continuum of cell states characterized by transcriptional changes in hormone-, growth factor-, and milk production-related pathways. Generalized additive models suggest that one subcluster, LC1 epithelial cells, increases as a function of time postpartum, daycare attendance, and the use of hormonal birth control. We identify several subclusters of macrophages in hBM that are enriched for tolerogenic functions, possibly playing a role in protecting the mammary gland during lactation. Our description of the cellular components of breast milk, their association with maternal–infant dyad metadata, and our quantification of alterations at the gene and pathway levels provide a detailed longitudinal picture of hBM cells across lactational time. This work paves the way for future investigations of how a potential division of cellular labor and differential hormone regulation might be leveraged therapeutically to support healthy lactation and potentially aid in milk production.
Collapse
Affiliation(s)
- Sarah K. Nyquist
- Broad Institute of MIT and Harvard, Cambridge, MA 02142
- Program in Computational and Systems Biology, Massachusetts Institute of Technology; Cambridge, MA 02139
- Department of Chemistry and Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139
- Computer Science and Artificial Intelligence Laboratory, Department of Mathematics, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Patricia Gao
- Department of Chemistry and Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Tessa K. J. Haining
- Department of Chemistry and Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Michael R. Retchin
- Department of Chemistry and Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Yarden Golan
- Department of Bioengineering and Therapeutic Sciences, Institute for Human Genetics, University of California, San Francisco, CA 94143
| | - Riley S. Drake
- Broad Institute of MIT and Harvard, Cambridge, MA 02142
- Department of Chemistry and Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139
| | - Kellie Kolb
- Broad Institute of MIT and Harvard, Cambridge, MA 02142
- Department of Chemistry and Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Benjamin E. Mead
- Broad Institute of MIT and Harvard, Cambridge, MA 02142
- Department of Chemistry and Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Nadav Ahituv
- Department of Bioengineering and Therapeutic Sciences, Institute for Human Genetics, University of California, San Francisco, CA 94143
| | | | - Alex K. Shalek
- Broad Institute of MIT and Harvard, Cambridge, MA 02142
- Program in Computational and Systems Biology, Massachusetts Institute of Technology; Cambridge, MA 02139
- Department of Chemistry and Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Division of Health Science & Technology, Harvard Medical School, Boston, MA 02115
- Department of Immunology, Massachusetts General Hospital, Boston, MA 02114
| | - Bonnie Berger
- Broad Institute of MIT and Harvard, Cambridge, MA 02142
- Computer Science and Artificial Intelligence Laboratory, Department of Mathematics, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Brittany A. Goods
- Thayer School of Engineering, Program in Quantitative Biomedical Sciences, Dartmouth College, Hanover, NH 03755
| |
Collapse
|
22
|
Jager S, Cramer DAT, Hoek M, Mokiem NJ, van Keulen BJ, van Goudoever JB, Dingess KA, Heck AJR. Proteoform Profiles Reveal That Alpha-1-Antitrypsin in Human Serum and Milk Is Derived From a Common Source. Front Mol Biosci 2022; 9:858856. [PMID: 35274008 PMCID: PMC8902301 DOI: 10.3389/fmolb.2022.858856] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 02/08/2022] [Indexed: 12/04/2022] Open
Abstract
The Alpha-1-Antitrypsin (A1AT) protein is an important protease inhibitor highly abundant in human serum and other body fluids. Additional to functioning as a protease inhibitor, A1AT is an important acute phase protein. Here, we set out to compare the proteoform profiles of A1AT purified from the human serum and milk of eight healthy donors to determine the origin of human milk A1AT. Following affinity purification, size-exclusion chromatography coupled to native mass spectrometry was used to monitor individual proteoform profiles comparing inter- and intra-donor profiles. The A1AT intra-donor proteoform profiles were found to be highly identical between serum and milk, while they were highly distinct between donors, even when comparing only serum or milk samples. The observed inter-donor proteoform variability was due to differences in the abundances of different N-glycoforms, mainly due to branching, fucosylation, and the relative abundance of N-terminally processed A1AT fragments. From our data we conclude that nearly all A1AT in serum and milk is synthesized by a common source, i.e. the liver, and then secreted into the circulation and enters the mammary gland via diffusion or transport. Thereby, proteoform profile changes, as seen upon infection and/or inflammation in the blood will be reflected in the milk, which may then be transferred to the breastfed infant.
Collapse
Affiliation(s)
- Shelley Jager
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
| | - Dario A. T. Cramer
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
| | - Max Hoek
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
| | - Nadia J. Mokiem
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
| | - Britt J. van Keulen
- Department of Pediatrics, Vrije Universiteit, University of Amsterdam Emma Children’s Hospital, Amsterdam UMC, Amsterdam, Netherlands
| | - Johannes B. van Goudoever
- Department of Pediatrics, Vrije Universiteit, University of Amsterdam Emma Children’s Hospital, Amsterdam UMC, Amsterdam, Netherlands
| | - Kelly A. Dingess
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
- Department of Pediatrics, Vrije Universiteit, University of Amsterdam Emma Children’s Hospital, Amsterdam UMC, Amsterdam, Netherlands
| | - Albert J. R. Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
- *Correspondence: Albert J. R. Heck,
| |
Collapse
|
23
|
Dekker PM, Boeren S, van Goudoever JB, Vervoort JJM, Hettinga KA. Exploring Human Milk Dynamics: Interindividual Variation in Milk Proteome, Peptidome, and Metabolome. J Proteome Res 2022; 21:1002-1016. [PMID: 35104145 PMCID: PMC8981310 DOI: 10.1021/acs.jproteome.1c00879] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
![]()
Human milk is a dynamic
biofluid, and its detailed composition
receives increasing attention. While most studies focus on changes
over time or differences between maternal characteristics, interindividual
variation receives little attention. Nevertheless, a comprehensive
insight into this can help interpret human milk studies and help human
milk banks provide targeted milk for recipients. This study aimed
to map interindividual variation in the human milk proteome, peptidome,
and metabolome and to investigate possible explanations for this variation.
A set of 286 milk samples was collected from 29 mothers in the third
month postpartum. Samples were pooled per mother, and proteins, peptides,
and metabolites were analyzed. A substantial coefficient of variation
(>100%) was observed for 4.6% and 36.2% of the proteins and peptides,
respectively. In addition, using weighted correlation network analysis
(WGCNA), 5 protein and 11 peptide clusters were obtained, showing
distinct characteristics. With this, several associations were found
between the different data sets and with specific sample characteristics.
This study provides insight into the dynamics of human milk protein,
peptide, and metabolite composition. In addition, it will support
future studies that evaluate the effect size of a parameter of interest
by enabling a comparison with natural variability.
Collapse
Affiliation(s)
- Pieter M Dekker
- Food Quality and Design Group, Wageningen University & Research, Bornse Weilanden 9, 6708 WG Wageningen, The Netherlands.,Laboratory of Biochemistry, Wageningen University & Research, Stippeneng 4, 6708 WE Wageningen, The Netherlands
| | - Sjef Boeren
- Laboratory of Biochemistry, Wageningen University & Research, Stippeneng 4, 6708 WE Wageningen, The Netherlands
| | - Johannes B van Goudoever
- Department of Pediatrics, Amsterdam UMC Vrije Universiteit Emma Children's Hospital, 1081 Amsterdam, The Netherlands
| | - Jacques J M Vervoort
- Laboratory of Biochemistry, Wageningen University & Research, Stippeneng 4, 6708 WE Wageningen, The Netherlands
| | - Kasper A Hettinga
- Food Quality and Design Group, Wageningen University & Research, Bornse Weilanden 9, 6708 WG Wageningen, The Netherlands
| |
Collapse
|
24
|
Gustafsson A, Johansson E, Henckel E, Lange S, Bohlin K. Changes in Antisecretory Factor in Human Milk During the Postpartum and Length of Gestation. J Hum Lact 2022; 38:131-140. [PMID: 34074170 PMCID: PMC8814942 DOI: 10.1177/08903344211021306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
BACKGROUND Preterm infants are more susceptible to inflammatory complications than term infants. Human milk contains numerous bioactive components protecting the newborn infant. Antisecretory factor, a protein regulating secretory and inflammatory processes by complex binding with complement factors, is present in human milk. RESEARCH AIMS To describe antisecretory factor (1) in mother's own milk in term and preterm infants; and (2) in donor milk before and after Holder pasteurization. METHODS The study was prospective, longitudinal, explorative, and descriptive. Antisecretory factor-compleasome was determined using sandwich enzyme-linked immunosorbent assay in longitudinal human milk samples over 12 weeks from mothers (N = 87) of term (n = 41) and of preterm (n = 46) infants and 20 anonymized donor human milk samples before and after Holder pasteurization. RESULTS Antisecretory factor-compleasome was overall higher in colostrum versus mature milk (p < .001) and no difference was found in term or preterm colostrum (p = .82). In mature milk, compleasome was higher and more variable in the preterm group (p = .01). After Holder pasteurization, compleasome levels increased (p < .001). CONCLUSION Antisecretory factor followed the pattern of other immunological factors with high levels in colostrum. After preterm birth, levels of antisecretory factor were higher and more variable in mature milk. Holder pasteurization did not degrade antisecretory factor, indicating preserved anti-inflammatory properties in donor human milk.
Collapse
Affiliation(s)
- Anna Gustafsson
- 27106 Department of Neonatology, Karolinska University Hospital, Stockholm, Sweden.,206106 Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Ewa Johansson
- 99018 Department of Infectious Diseases, Institute of Biomedicine, Gothenburg University, Gothenburg, Sweden.,Department of Clinical Microbiology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Ewa Henckel
- 27106 Department of Neonatology, Karolinska University Hospital, Stockholm, Sweden.,206106 Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Stefan Lange
- 99018 Department of Infectious Diseases, Institute of Biomedicine, Gothenburg University, Gothenburg, Sweden.,Department of Clinical Microbiology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Kajsa Bohlin
- 27106 Department of Neonatology, Karolinska University Hospital, Stockholm, Sweden.,206106 Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
25
|
Bondt A, Dingess KA, Hoek M, van Rijswijck DMH, Heck AJR. A Direct MS-Based Approach to Profile Human Milk Secretory Immunoglobulin A (IgA1) Reveals Donor-Specific Clonal Repertoires With High Longitudinal Stability. Front Immunol 2021; 12:789748. [PMID: 34938298 PMCID: PMC8685336 DOI: 10.3389/fimmu.2021.789748] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/17/2021] [Indexed: 12/29/2022] Open
Abstract
Recently, a mass spectrometry-based approach was introduced to directly assess the IgG1 immunoglobulin clonal repertoires in plasma. Here we expanded upon this approach by describing a mass spectrometry-based technique to assess specifically the clonal repertoire of another important class of immunoglobulin molecules, IgA1, and show it is efficiently and robustly applicable to either milk or plasma samples. Focusing on two individual healthy donors, whose milk was sampled longitudinally during the first 16 weeks of lactation, we demonstrate that the total repertoire of milk sIgA1 is dominated by only 50-500 clones, even though the human body theoretically can generate several orders of magnitude more clones. We show that in each donor the sIgA1 repertoire only changes marginally and quite gradually over the monitored 16-week period of lactation. Furthermore, the observed overlap in clonal repertoires between the two individual donors is close to non-existent. Mothers provide protection to their newborn infants directly by the transfer of antibodies via breastfeeding. The approach introduced here, can be used to visualize the clonal repertoire transferred from mother to infant and to detect changes in-time in that repertoire adapting to changes in maternal physiology.
Collapse
Affiliation(s)
- Albert Bondt
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Netherlands Proteomics Center, Utrecht, Netherlands
| | - Kelly A Dingess
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Netherlands Proteomics Center, Utrecht, Netherlands
| | - Max Hoek
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Netherlands Proteomics Center, Utrecht, Netherlands
| | - Danique M H van Rijswijck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Netherlands Proteomics Center, Utrecht, Netherlands
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Netherlands Proteomics Center, Utrecht, Netherlands
| |
Collapse
|
26
|
Analysis of the Endogenous Peptidomes of Different Infant Formula Types and Human Milk. Foods 2021; 10:foods10112579. [PMID: 34828867 PMCID: PMC8623676 DOI: 10.3390/foods10112579] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/15/2021] [Accepted: 10/22/2021] [Indexed: 01/06/2023] Open
Abstract
Infant formula (IF) is a commonly used replacement whenever mother’s own milk is not available. Most IFs are based on cow milk (powders, liquids). Alternatives, based on other sources such as goat milk or plants, exist. Independent of the source, IF production and composition are strictly regulated. Besides proteins, minerals, and lipids, milk contains a variety of endogenous peptides. Whereas the human milk peptidome has been studied intensively, the peptidomes of IFs have been mostly neglected. This study investigated the peptidomes of different types of first stage IF, including cow milk-based powders and liquids, and powdered goat milk-based IF, highlighting major similarities and differences to human milk. Extracted native peptidomes were analyzed by nanoRPC-ESI-MS/MS using two different fragmentation techniques allowing the confident identification of 1587 peptides. β-Casein peptides dominated in all samples. Interestingly, powdered and liquid cow milk-based IFs differed in the numbers of β- and αS1-casein peptides, indicating processing-derived variations. However, the peptidomes of cow and goat milk-based IF appeared to be more comparable to each other than to human milk. Despite an overlap in the major source proteins, many peptide sequences were different, i.e., species-specific. Remarkably, the data indicate that the human milk peptidome might be donor-specific as well.
Collapse
|
27
|
Dingess KA, Gazi I, van den Toorn HWP, Mank M, Stahl B, Reiding KR, Heck AJR. Monitoring Human Milk β-Casein Phosphorylation and O-Glycosylation Over Lactation Reveals Distinct Differences between the Proteome and Endogenous Peptidome. Int J Mol Sci 2021; 22:8140. [PMID: 34360914 PMCID: PMC8347866 DOI: 10.3390/ijms22158140] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 12/15/2022] Open
Abstract
Human milk is a vital biofluid containing a myriad of molecular components to ensure an infant's best start at a healthy life. One key component of human milk is β-casein, a protein which is not only a structural constituent of casein micelles but also a source of bioactive, often antimicrobial, peptides contributing to milk's endogenous peptidome. Importantly, post-translational modifications (PTMs) like phosphorylation and glycosylation typically affect the function of proteins and peptides; however, here our understanding of β-casein is critically limited. To uncover the scope of proteoforms and endogenous peptidoforms we utilized mass spectrometry (LC-MS/MS) to achieve in-depth longitudinal profiling of β-casein from human milk, studying two donors across 16 weeks of lactation. We not only observed changes in β-casein's known protein and endogenous peptide phosphorylation, but also in previously unexplored O-glycosylation. This newly discovered PTM of β-casein may be important as it resides on known β-casein-derived antimicrobial peptide sequences.
Collapse
Affiliation(s)
- Kelly A. Dingess
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, 3584 CH Utrecht, The Netherlands; (K.A.D.); (I.G.); (H.W.P.v.d.T.)
- Netherlands Proteomics Center, 3584 CH Utrecht, The Netherlands
| | - Inge Gazi
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, 3584 CH Utrecht, The Netherlands; (K.A.D.); (I.G.); (H.W.P.v.d.T.)
- Netherlands Proteomics Center, 3584 CH Utrecht, The Netherlands
| | - Henk W. P. van den Toorn
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, 3584 CH Utrecht, The Netherlands; (K.A.D.); (I.G.); (H.W.P.v.d.T.)
- Netherlands Proteomics Center, 3584 CH Utrecht, The Netherlands
| | - Marko Mank
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (M.M.); (B.S.)
| | - Bernd Stahl
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (M.M.); (B.S.)
- Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, 3584 CG Utrecht, The Netherlands
| | - Karli R. Reiding
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, 3584 CH Utrecht, The Netherlands; (K.A.D.); (I.G.); (H.W.P.v.d.T.)
- Netherlands Proteomics Center, 3584 CH Utrecht, The Netherlands
| | - Albert J. R. Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, 3584 CH Utrecht, The Netherlands; (K.A.D.); (I.G.); (H.W.P.v.d.T.)
- Netherlands Proteomics Center, 3584 CH Utrecht, The Netherlands
| |
Collapse
|
28
|
Jia Q, Wang Y, Zhu J, Yu H, Tong X. A literature review on lactopontin and its roles in early life. Transl Pediatr 2021; 10:1924-1931. [PMID: 34430441 PMCID: PMC8349962 DOI: 10.21037/tp-21-293] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 07/14/2021] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE Our study aims to review the functions and possible mechanisms of lactopontin (LPN) in early life. BACKGROUND Human milk proteins provide a variety of protection and health benefits in early life. One of these multifunctional proteins is LPN, which is osteopontin (OPN) derived from milk. METHODS Information used to write this paper was collected from Uniprot, PubMed, and Google Scholar, including in vitro, in vivo, and clinical studies. CONCLUSIONS LPN is a highly phosphorylated, O-glycosylated acidic protein and a unique type of OPN, as it presents at the highest concentration and a higher degree of posttranslational modifications (PTMs) in human milk than other tissues and excretions. LPN is present in milk and the intestinal tracts of infants after consumption as a mixture of intact protein and peptides, which can bind diverse integrin and receptors in the target cell and drive downstream signaling pathways. LPN is found to play important roles in developing the immune, intestinal and nervous systems in early life. Moreover, LPN has also shown to support preterm infants' health when they are especially vulnerable after delivery via animal studies. Additionally, LPN can form protein complex with another milk bioactive protein, lactoferrin (LF), to withstand proteolysis and perform more efficient biological activity. Therefore, LPN showed great potential for early life while more clinical trials and evidence are still emergying.
Collapse
Affiliation(s)
- Qiong Jia
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| | - Yiran Wang
- Department of Nutritional and Functional Assessment, Beijing Institute of Nutritional Resources, Beijing, China
| | - Jing Zhu
- Department of Nutritional and Functional Assessment, Beijing Institute of Nutritional Resources, Beijing, China
| | - Huanling Yu
- Department of Nutrition and Food Hygiene, School of Public Health, Capital Medical University, Beijing, China
| | - Xiaomei Tong
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| |
Collapse
|
29
|
Optimization of a human milk-directed quantitative sIgA ELISA method substantiated by mass spectrometry. Anal Bioanal Chem 2021; 413:5037-5049. [PMID: 34169348 PMCID: PMC8405464 DOI: 10.1007/s00216-021-03468-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/27/2021] [Accepted: 06/09/2021] [Indexed: 12/17/2022]
Abstract
Immunoglobulins are the primary protective products in human milk and are responsible for transferring maternal pathogen memory to the infant, providing protection by binding to recognized pathogens and inhibiting virulence. To better understand potentially protective/anti-infective compounds in human milk, the establishment of human milk–tailored analytical approaches is crucial, as most contemporary analytical methods have been optimized for plasma or serum. One of the most prominent immunoglobulins in human milk is secretory immunoglobulin A (sIgA), which may be relevant for the protection of breastfed infants from harmful pathogens. Advanced sIgA detection methods can help monitor the immune status and development of the mother-infant dyad. We therefore developed an enzyme-linked immunosorbent assay (ELISA) sIgA method for the quantitative analysis of IgA plus secretory component (SC), validated with sIgA standards and substantiated by mass spectrometry (MS)–based proteomics. A very strong correlation was observed between the MS-detected IgA1 and the human milk–specific sIgA ELISA (r = 0.82). Overall, the MS data indicate that the developed human milk sIgA ELISA does not differentiate between sIgA1 and sIgA2 and is, therefore, a reflection of total sIgA. Furthermore, our MS data and the human milk–derived sIgA ELISA data are better correlated than data derived from a standard serum IgA ELISA kit (relative to MS IgA1 r = 0.82 and r = 0.42, respectively). We therefore propose our human milk–specific sIgA ELISA as an ideal quantitative indicator of total sIgA with advantages over current serum IgA ELISA kits.
Collapse
|