1
|
Rold LS, Jensen AM, Arenholt L, Leutscher PDC, Ovesen PG, Hagstrøm S, Sørensen S. Identifying microbiome-based changes and biomarkers prior to disease development in mother and child, with a focus on gestational diabetes mellitus: protocol for the DANish Maternal and Offspring Microbiome (DANMOM) cohort study. BMJ Open 2024; 14:e083358. [PMID: 39242166 PMCID: PMC11381651 DOI: 10.1136/bmjopen-2023-083358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 08/23/2024] [Indexed: 09/09/2024] Open
Abstract
INTRODUCTION The human gut microbiota is associated with gestational diabetes mellitus (GDM), which imposes a risk of developing long-term health problems for mother and child. Most studies on GDM and microbiota have been cross-sectional, which makes it difficult to make any conclusions on causality. Furthermore, it is important to assess if a dysbiotic microbiota is passed from the mother to the child, and then being at risk of developing metabolic health problems later in life. The DANish Maternal and Offspring Microbiome study aims to identify gut microbiota-related factors involved in metabolic dysfunction in women with GDM and their offspring. Importantly, the study design allows for early detection of biological changes associated with later development of metabolic disease. This could provide us with unique tools to support early diagnosis or implement preventative measures. METHODS AND ANALYSIS Pregnant women are included in the study after the 11-14 weeks' prenatal ultrasound scan and followed throughout pregnancy with enrolment of the offspring at birth. 202 women and 112 children have been included from North Denmark Regional Hospital and Aalborg University Hospital in Denmark. Mother and child are followed until the children reach the age of 5 years. From the mother, we collect faeces, urine, blood, saliva, vaginal fluid and breast milk samples, in addition to faeces and a blood sample from the child. Microbiota composition in biological samples will be analysed using 16S rRNA gene sequencing and compared with demographic and clinical data from medical charts, registers and questionnaires. Sample and data collection will continue until July 2028. ETHICS AND DISSEMINATION The study protocol has been approved by the North Denmark Region Committee on Health Research Ethics (N20190007). Written informed consent is obtained from all participants prior to study participation. Study results will be published in international peer-reviewed journals and presented at international conferences. The results will also be presented to the funders of the study and study participants.
Collapse
Affiliation(s)
- Louise Søndergaard Rold
- Centre for Clinical Research, North Denmark Regional Hospital, Hjørring, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Ann-Maria Jensen
- Centre for Clinical Research, North Denmark Regional Hospital, Hjørring, Denmark
| | - Louise Arenholt
- Centre for Clinical Research, North Denmark Regional Hospital, Hjørring, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Department of Gynecology and Obstetrics, North Denmark Regional Hospital, Hjørring, Denmark
| | - Peter Derek Christian Leutscher
- Centre for Clinical Research, North Denmark Regional Hospital, Hjørring, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Per Glud Ovesen
- Department of Gynecology and Obstetrics, Aarhus University Hospital, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus, Denmark
| | - Søren Hagstrøm
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Department of Pediatrics and Adolescent Medicine, Aalborg University Hospital, Aalborg, Denmark
- Steno Diabetes Center North Denmark, Aalborg, Denmark
| | - Suzette Sørensen
- Centre for Clinical Research, North Denmark Regional Hospital, Hjørring, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Steno Diabetes Center North Denmark, Aalborg, Denmark
| |
Collapse
|
2
|
Rold LS, Guldbæk JM, Lindegaard CS, Kirk S, Nygaard LD, Bundgaard-Nielsen C, Holm-Jacobsen JN, Leutscher P, Viuff ACF, Hagstrøm S, Sørensen S. A comparison of the breast milk microbiota from women diagnosed with gestational diabetes mellitus and women without gestational diabetes mellitus. BMC Pregnancy Childbirth 2024; 24:412. [PMID: 38849751 PMCID: PMC11157733 DOI: 10.1186/s12884-024-06604-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 05/22/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Human breast milk (HBM) is a contributing factor in modulating the infant's gut microbiota, as it contains bacteria that are directly transferred to the infant during breastfeeding. It has been shown that children of women diagnosed with gestational diabetes mellitus (GDM) have a different gut microbiota compared to children of women without GDM. Our hypothesis is therefore that women with GDM have a different HBM microbiota, which may influence the metabolic function and capacity of the child later in life. The aim of this study was to investigate whether women with GDM have a different breast milk microbiota 1-3 weeks postpartum compared to women without GDM. METHODS In this case-control study, a total of 45 women were included: 18 women with GDM and 27 women without GDM. A milk sample was collected from each participant 1 to 3 weeks postpartum and the bacterial composition was examined by 16 S rRNA gene sequencing targeting the V4 region. RESULTS High relative abundances of Streptococcus and Staphylococcus were present in samples from both women with and without GDM. No difference could be seen in either alpha diversity, beta diversity, or specific taxa between groups. CONCLUSION Our results did not support the existence of a GDM-associated breast milk microbiota at 1-3 weeks postpartum. Further research is needed to fully understand the development of the gut microbiota of infants born to mothers with GDM.
Collapse
Affiliation(s)
- Louise Søndergaard Rold
- Centre for Clinical Research, North Denmark Regional Hospital, Hjørring, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Steno Diabetes Center North Denmark, Aalborg, Denmark
| | | | | | - Stine Kirk
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | | | | | | | - Peter Leutscher
- Centre for Clinical Research, North Denmark Regional Hospital, Hjørring, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | | | - Søren Hagstrøm
- Centre for Clinical Research, North Denmark Regional Hospital, Hjørring, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Steno Diabetes Center North Denmark, Aalborg, Denmark
- Department of Pediatrics and Adolescents, Aalborg University Hospital, Aalborg, Denmark
| | - Suzette Sørensen
- Centre for Clinical Research, North Denmark Regional Hospital, Hjørring, Denmark.
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.
- Steno Diabetes Center North Denmark, Aalborg, Denmark.
| |
Collapse
|
3
|
Jiang H, Xu Y, Chen G, Liu T, Yang Y, Mao X. Digestive properties and peptide profiles exhibited significant differences between skim camel milk and bovine milk powder after static in vitro simulated infant gastrointestinal digestion. Food Res Int 2024; 178:113860. [PMID: 38309893 DOI: 10.1016/j.foodres.2023.113860] [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: 10/12/2023] [Revised: 12/09/2023] [Accepted: 12/14/2023] [Indexed: 02/05/2024]
Abstract
This study aims to analyze the differences in digestion properties and peptide profiles between the skim camel and bovine milk powder after static in vitro simulated infant gastrointestinal digestion. The hydrolysis degree of camel milk proteins exceeded by 13.18% that of bovine milk. The concentration and release rate of free amino groups in the camel milk digesta was higher than that of bovine milk powder, which was likely due to the higher β-/αs-casein ratio and larger casein micelle size in camel milk. Camel milk powder presented higher β-CN coverage and comparatively shorter bioactive peptides compared to bovine milk powder. The anti-inflammatory peptide KVLPVPQ displayed the highest abundance in camel milk powder. Outcomes of this study showed that camel milk proteins possessed superior digestibility and unique peptides, which outlined the potential nutritional implications of camel milk for infants.
Collapse
Affiliation(s)
- Hui Jiang
- Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Yunxuan Xu
- Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Gangliang Chen
- Xinjiang Wangyuan Camel Milk Industrial Co., Ltd., Altay 836400, China
| | - Tianchong Liu
- Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Yitong Yang
- Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Xueying Mao
- Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| |
Collapse
|
4
|
Ramos-Tapia I, Reynaldos-Grandón KL, Pérez-Losada M, Castro-Nallar E. Characterization of the upper respiratory tract microbiota in Chilean asthmatic children reveals compositional, functional, and structural differences. FRONTIERS IN ALLERGY 2023; 4:1223306. [PMID: 37577334 PMCID: PMC10419220 DOI: 10.3389/falgy.2023.1223306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/20/2023] [Indexed: 08/15/2023] Open
Abstract
Around 155 million people worldwide suffer from asthma. In Chile, the prevalence of this disease in children is around 15% and has a high impact in the health system. Studies suggest that asthma is caused by multiple factors, including host genetics, antibiotic use, and the development of the airway microbiota. Here, we used 16S rRNA high-throughput sequencing to characterize the nasal and oral mucosae of 63 asthmatic and 89 healthy children (152 samples) from Santiago, Chile. We found that the nasal mucosa was dominated by a high abundance of Moraxella, Dolosigranulum, Haemophilus, Corynebacterium, Streptococcus, and Staphylococcus. In turn, the oral mucosa was characterized by a high abundance of Streptococcus, Haemophilus, Gemella, Veillonella, Neisseria, and Porphyromonas. Our results showed significantly (P < 0.001) lower alpha diversity and an over-abundance of Streptococcus (P < 0.01) in nasal samples from asthmatics compared to samples from healthy subjects. Community structure, as revealed by co-occurrence networks, showed different microbial interactions in asthmatic and healthy subjects, particularly in the nasal microbiota. The networks revealed keystone genera in each body site, including Prevotella, Leptotrichia, and Porphyromonas in the nasal microbiota, and Streptococcus, Granulicatella, and Veillonella in the oral microbiota. We also detected 51 functional pathways differentially abundant on the nasal mucosa of asthmatic subjects, although only 13 pathways were overrepresented in the asthmatic subjects (P < 0.05). We did not find any significant differences in microbial taxonomic (composition and structure) and functional diversity between the oral mucosa of asthmatic and healthy subjects. This study explores for the first time the relationships between the upper respiratory airways bacteriome and asthma in Chile. It demonstrates that the nasal cavity of children from Santiago harbors unique bacterial communities and identifies potential taxonomic and functional biomarkers of pediatric asthma.
Collapse
Affiliation(s)
- Ignacio Ramos-Tapia
- Centro de Bioinformática y Biología Integrativa, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | | | - Marcos Pérez-Losada
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, The George Washington University, Washington, DC, United States
| | - Eduardo Castro-Nallar
- Departamento de Microbiología, Facultad de Ciencias de la Salud, Universidad de Talca, Talca, Chile
- Centro de Ecología Integrativa, Universidad de Talca, Talca, Chile
| |
Collapse
|
5
|
Koivusaari K, Niinistö S, Nevalainen J, Honkanen J, Ruohtula T, Koreasalo M, Ahonen S, Åkerlund M, Tapanainen H, Siljander H, Miettinen ME, Alatossava T, Ilonen J, Vaarala O, Knip M, Virtanen SM. Infant Feeding, Gut Permeability, and Gut Inflammation Markers. J Pediatr Gastroenterol Nutr 2023; 76:822-829. [PMID: 36913717 DOI: 10.1097/mpg.0000000000003756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/15/2023]
Abstract
OBJECTIVES Increased gut permeability and gut inflammation have been linked to the development of type 1 diabetes. Little is known on whether and how intake of different foods is linked to these mechanisms in infancy. We investigated whether the amount of breast milk and intake of other foods are associated with gut inflammation marker concentrations and permeability. METHODS Seventy-three infants were followed from birth to 12 months of age. Their diet was assessed with structured questionnaires and 3-day weighed food records at the age of 3, 6, 9, and 12 months. Gut permeability was assessed with the lactulose/mannitol test and fecal calprotectin and human β-defensin-2 (HBD-2) concentrations were analyzed from stool samples at the age of 3, 6, 9, and 12 months. The associations between foods and gut inflammation marker concentrations and permeability were analyzed using generalized estimating equations. RESULTS Gut permeability and gut inflammation marker concentrations decreased during the first year of life. Intake of hydrolyzed infant formula ( P = 0.003) and intake of fruits and juices ( P = 0.001) were associated with lower intestinal permeability. Intake of fruits and juices ( P < 0.001), vegetables ( P < 0.001), and oats ( P = 0.003) were associated with lower concentrations of HBD-2. Higher intake of breast milk was associated with higher fecal calprotectin concentrations ( P < 0.001), while intake of fruits and juices ( P < 0.001), vegetables ( P < 0.001), and potatoes ( P = 0.007) were associated with lower calprotectin concentrations. CONCLUSIONS Higher intake of breast milk may contribute to higher calprotectin concentration, whereas several complementary foods may decrease gut permeability and concentrations of calprotectin and HBD-2 in infant gut.
Collapse
Affiliation(s)
- Katariina Koivusaari
- From the Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- the Department of Food and Nutrition, University of Helsinki, Helsinki, Finland
| | - Sari Niinistö
- From the Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Jaakko Nevalainen
- the Faculty of Social Sciences, Unit of Health Sciences, Tampere University, Tampere, Finland
| | - Jarno Honkanen
- the Research Program for Translational Immunology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Terhi Ruohtula
- the Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mirva Koreasalo
- From the Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Suvi Ahonen
- From the Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Mari Åkerlund
- From the Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Heli Tapanainen
- From the Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Heli Siljander
- From the Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Maija E Miettinen
- From the Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Tapani Alatossava
- the Department of Food and Nutrition, University of Helsinki, Helsinki, Finland
| | - Jorma Ilonen
- the Immunogenetics Laboratory, University of Turku, Turku, Finland
| | - Outi Vaarala
- the Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mikael Knip
- From the Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Suvi M Virtanen
- From the Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| |
Collapse
|
6
|
Bednarczyk MM, Żelazna O, Głuszczak‐Idziakowska E, Schreiber‐Zamora J, Furmanek M, Kociszewska‐Najman B. A case of necrotizing enterocolitis in full-term infant on his first day of life-Early presentation with multifactorial etiology. Clin Case Rep 2023; 11:e7150. [PMID: 37096169 PMCID: PMC10122096 DOI: 10.1002/ccr3.7150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/20/2023] [Accepted: 03/09/2023] [Indexed: 04/26/2023] Open
Abstract
We aimed to show the complexity of NEC in the full-term newborns. Additionally, we wanted to underline the importance of breastfeeding from the first day of life and show how deteriorating effect the antibiotics can have on gastrointestinal tract.
Collapse
Affiliation(s)
| | - Olga Żelazna
- Student Scientific Group at the Department Neonatology “Proneo”Medical University of WarsawWarsawPoland
| | | | | | - Mariusz Furmanek
- Department of Pediatric RadiologyMedical University of WarsawWarsawPoland
| | | |
Collapse
|
7
|
Sakurai K, Yamamoto M, Eguchi A, Takatani R, Watanabe M, Mori C. Association between maternal antibiotic exposure during pregnancy and childhood obesity in the Japan Environment and Children's Study. Pediatr Obes 2022; 17:e12956. [PMID: 35751177 PMCID: PMC9787574 DOI: 10.1111/ijpo.12956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 04/20/2022] [Accepted: 06/06/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND The association between maternal antibiotic exposure during pregnancy and childhood obesity is still unclear. OBJECTIVES The study aimed to evaluate the association between prenatal exposure to antibiotics and obesity at age 3 years using data from a large Japanese birth cohort. METHODS The Japan Environment and Children's Study is a nationwide birth cohort study. In this study, singleton vaginal full-term births were included. Obesity was defined as body mass index ≥95th percentile according to child growth standards. Prenatal antibiotic exposure was defined as antimicrobial agent use during pregnancy and was collected from maternal interviews and medical record transcripts. Logistic regression analysis was performed to evaluate the association of prenatal antibiotic exposure with child obesity at 3 years. RESULTS In the crude and adjusted models with all children, maternal antibiotic exposure during pregnancy showed a marginal relationship with child obesity at 3 years. In the analyses according to exposure period and sex, exposure to antibiotics during the second/third trimester was significantly associated with obesity at the age of 3 years in female infants, but not in male infants, although the exposure during the first trimester was not in both sexes. CONCLUSION Maternal antibiotic exposure during mid/late pregnancy may result in child obesity.
Collapse
Affiliation(s)
- Kenichi Sakurai
- Department of Nutrition and Metabolic Medicine, Center for Preventive Medical SciencesChiba UniversityChibaJapan
| | - Midori Yamamoto
- Department of Sustainable Health Science, Center for Preventive Medical SciencesChiba UniversityChibaJapan
| | - Akifumi Eguchi
- Department of Sustainable Health Science, Center for Preventive Medical SciencesChiba UniversityChibaJapan
| | - Rieko Takatani
- Department of Sustainable Health Science, Center for Preventive Medical SciencesChiba UniversityChibaJapan
| | - Masahiro Watanabe
- Department of Sustainable Health Science, Center for Preventive Medical SciencesChiba UniversityChibaJapan
| | - Chisato Mori
- Department of Sustainable Health Science, Center for Preventive Medical SciencesChiba UniversityChibaJapan,Department of Bioenvironmental Medicine, Graduate School of MedicineChiba UniversityChibaJapan
| | | |
Collapse
|
8
|
Yuan Q, Gong H, Du M, Li T, Mao X. Milk fat globule membrane supplementation to obese rats during pregnancy and lactation promotes neurodevelopment in offspring via modulating gut microbiota. Front Nutr 2022; 9:945052. [PMID: 36046136 PMCID: PMC9421050 DOI: 10.3389/fnut.2022.945052] [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] [Received: 05/16/2022] [Accepted: 07/18/2022] [Indexed: 12/12/2022] Open
Abstract
Pre-pregnancy obesity and high-fat diet (HFD) during pregnancy and lactation are associated with neurodevelopmental delay in offspring. This study aimed to investigate whether milk fat globule membrane (MFGM) supplementation in obese dams could promote neurodevelopment in offspring. Obese female rats induced by HFD were supplemented with MFGM during pregnancy and lactation. Maternal HFD exposure significantly delayed the maturation of neurological reflexes and inhibited neurogenesis in offspring, which were significantly recovered by maternal MFGM supplementation. Gut microbiota analysis revealed that MFGM supplementation modulated the diversity and composition of gut microbiota in offspring. The abundance of pro-inflammatory bacteria such as Escherichia shigella and Enterococcus were down-regulated, and the abundance of bacteria with anti-inflammatory and anti-obesity functions, such as Akkermansia and Lactobacillus were up-regulated. Furthermore, MFGM alleviated neuroinflammation by decreasing the levels of lipopolysaccharides (LPS) and pro-inflammatory cytokines in the circulation and brain, as well as inhibiting the activation of microglia. Spearman’s correlation analysis suggested that there existed a correlation between gut microbiota and inflammation-related indexes. In conclusion, maternal MFGM supplementation promotes neurodevelopment partly via modulating gut microbiota in offspring.
Collapse
Affiliation(s)
- Qichen Yuan
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, Ministry of Education, China Agricultural University, Beijing, China
| | - Han Gong
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, Ministry of Education, China Agricultural University, Beijing, China
| | - Min Du
- Department of Animal Sciences, Washington State University, Pullman, WA, United States
| | - Tiange Li
- Henan Engineering Technology Research Center of Food Processing and Circulation Safety Control, College of Food Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Xueying Mao
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, Ministry of Education, China Agricultural University, Beijing, China
| |
Collapse
|
9
|
Burge K, Eckert J, Wilson A, Trammell M, Lueschow SR, McElroy SJ, Dyer D, Chaaban H. Hyaluronic Acid 35 kDa Protects against a Hyperosmotic, Formula Feeding Model of Necrotizing Enterocolitis. Nutrients 2022; 14:nu14091779. [PMID: 35565748 PMCID: PMC9105773 DOI: 10.3390/nu14091779] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/20/2022] [Accepted: 04/22/2022] [Indexed: 12/23/2022] Open
Abstract
Necrotizing enterocolitis (NEC), an inflammatory disease of the intestine, is a common gastrointestinal emergency among preterm infants. Intestinal barrier dysfunction, hyperactivation of the premature immune system, and dysbiosis are thought to play major roles in the disease. Human milk (HM) is protective, but the mechanisms underpinning formula feeding as a risk factor in the development of NEC are incompletely understood. Hyaluronic acid 35 kDa (HA35), a bioactive glycosaminoglycan of HM, accelerates intestinal development in murine pups during homeostasis. In addition, HA35 prevents inflammation-induced tissue damage in pups subjected to murine NEC, incorporating Paneth cell dysfunction and dysbiosis. We hypothesized HA35 treatment would reduce histological injury and mortality in a secondary mouse model of NEC incorporating formula feeding. NEC-like injury was induced in 14-day mice by dithizone-induced disruption of Paneth cells and oral gavage of rodent milk substitute. Mortality and histological injury, serum and tissue cytokine levels, stool bacterial sequencing, and bulk RNA-Seq comparisons were analyzed. HA35 significantly reduced the severity of illness in this model, with a trend toward reduced mortality, while RNA-Seq analysis demonstrated HA35 upregulated genes associated with goblet cell function and innate immunity. Activation of these critical protective and reparative mechanisms of the small intestine likely play a role in the reduced pathology and enhanced survival trends of HA-treated pups subjected to intestinal inflammation in this secondary model of NEC, providing potentially interesting translational targets for the human preterm disease.
Collapse
Affiliation(s)
- Kathryn Burge
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.B.); (J.E.); (A.W.)
| | - Jeffrey Eckert
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.B.); (J.E.); (A.W.)
| | - Adam Wilson
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.B.); (J.E.); (A.W.)
| | - MaJoi Trammell
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (M.T.); (D.D.)
| | - Shiloh R. Lueschow
- Department of Microbiology and Immunology, Stead Family Department of Pediatrics, University of Iowa, Iowa City, IA 52242, USA;
| | - Steven J. McElroy
- Department of Pediatrics, University of California Davis, Sacramento, CA 95817, USA;
| | - David Dyer
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (M.T.); (D.D.)
| | - Hala Chaaban
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.B.); (J.E.); (A.W.)
- Correspondence:
| |
Collapse
|
10
|
Wang ZW, Gao YN, Huang SN, Wang JQ, Zheng N. Ex Vivo and In Vitro Studies Revealed Underlying Mechanisms of Immature Intestinal Inflammatory Responses Caused by Aflatoxin M1 Together with Ochratoxin A. Toxins (Basel) 2022; 14:toxins14030173. [PMID: 35324670 PMCID: PMC8953104 DOI: 10.3390/toxins14030173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/08/2022] [Accepted: 02/15/2022] [Indexed: 12/04/2022] Open
Abstract
Aflatoxin M1 (AFM1) and ochratoxin A (OTA), which are occasionally detected in milk and commercial baby foods, could easily enter and reach the gastrointestinal tract, posing impairment to the first line of defense and causing dysfunction of the tissue. The objective of this study was to investigate the immunostimulatory roles of individual and combined AFM1 and OTA on the immature intestine. Thus, we used ELISA assays to evaluate the generation of cytokines from ex vivo CD-1 fetal mouse jejunum induced by AFM1 and OTA and explored the related regulatory pathways and pivot genes using RNA-seq analysis. It was found that OTA exhibited much stronger ability in stimulating pro-inflammatory cytokine IL-6 from jejunum tissues than AFM1 (OTA of 4 μM versus AFM1 of 50 μM), whereas the combination of the two toxins seemed to exert antagonistic actions. In addition, transcriptomics also showed that most gene members in the enriched pathway ‘cytokine–cytokine receptor interaction’ were more highly expressed in OTA than the AFM1 group. By means of PPI network analysis, NFKB1 and RelB were regarded as hub genes in response to OTA but not AFM1. In the human FHs 74 Int cell line, both AFM1 and OTA enhanced the content of reactive oxygen species, and the oxidative response was more apparent in OTA-treated cells in comparison with AFM1. Furthermore, OTA and AFM1 + OTA raised the protein abundance of p50/RelB, and triggered the translocation of the dimer from cytosol to nucleus. Therefore, the experimental data ex vivo and in vitro showed that OTA-induced inflammation was thought to be bound up with the up-regulation and translocation of NF-κB, though AFM1 seemed to have no obvious impact. Since it was the first attempt to uncover the appearances and inner mechanisms regarding inflammation provoked by AFM1 and OTA on immature intestinal models, further efforts are needed to understand the detailed metabolic steps of the toxin in cells and to clarify their causal relationship with the signals proposed from current research.
Collapse
Affiliation(s)
- Zi-Wei Wang
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Z.-W.W.); (Y.-N.G.); (S.-N.H.); (J.-Q.W.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Milk and Milk Products Inspection Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Ya-Nan Gao
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Z.-W.W.); (Y.-N.G.); (S.-N.H.); (J.-Q.W.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Milk and Milk Products Inspection Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Sheng-Nan Huang
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Z.-W.W.); (Y.-N.G.); (S.-N.H.); (J.-Q.W.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Milk and Milk Products Inspection Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Jia-Qi Wang
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Z.-W.W.); (Y.-N.G.); (S.-N.H.); (J.-Q.W.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Milk and Milk Products Inspection Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Nan Zheng
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Z.-W.W.); (Y.-N.G.); (S.-N.H.); (J.-Q.W.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Milk and Milk Products Inspection Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Correspondence:
| |
Collapse
|
11
|
George S, Aguilera X, Gallardo P, Farfán M, Lucero Y, Torres JP, Vidal R, O'Ryan M. Bacterial Gut Microbiota and Infections During Early Childhood. Front Microbiol 2022; 12:793050. [PMID: 35069488 PMCID: PMC8767011 DOI: 10.3389/fmicb.2021.793050] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/10/2021] [Indexed: 12/12/2022] Open
Abstract
Gut microbiota composition during the first years of life is variable, dynamic and influenced by both prenatal and postnatal factors, such as maternal antibiotics administered during labor, delivery mode, maternal diet, breastfeeding, and/or antibiotic consumption during infancy. Furthermore, the microbiota displays bidirectional interactions with infectious agents, either through direct microbiota-microorganism interactions or indirectly through various stimuli of the host immune system. Here we review these interactions during childhood until 5 years of life, focusing on bacterial microbiota, the most common gastrointestinal and respiratory infections and two well characterized gastrointestinal diseases related to dysbiosis (necrotizing enterocolitis and Clostridioides difficile infection). To date, most peer-reviewed studies on the bacterial microbiota in childhood have been cross-sectional and have reported patterns of gut dysbiosis during infections as compared to healthy controls; prospective studies suggest that most children progressively return to a "healthy microbiota status" following infection. Animal models and/or studies focusing on specific preventive and therapeutic interventions, such as probiotic administration and fecal transplantation, support the role of the bacterial gut microbiota in modulating both enteric and respiratory infections. A more in depth understanding of the mechanisms involved in the establishment and maintenance of the early bacterial microbiota, focusing on specific components of the microbiota-immunity-infectious agent axis is necessary in order to better define potential preventive or therapeutic tools against significant infections in children.
Collapse
Affiliation(s)
- Sergio George
- Host-Pathogen Interaction Laboratory, Microbiology and Mycology Program, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Ximena Aguilera
- School of Medicine, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Pablo Gallardo
- Department of Pediatrics and Pediatric Surgery, Dr. Luis Calvo Mackenna Hospital, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Mauricio Farfán
- Department of Pediatrics and Pediatric Surgery, Dr. Luis Calvo Mackenna Hospital, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Yalda Lucero
- Host-Pathogen Interaction Laboratory, Microbiology and Mycology Program, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile.,Department of Pediatrics and Pediatric Surgery, Dr. Roberto del Río Hospital, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Juan Pablo Torres
- Host-Pathogen Interaction Laboratory, Microbiology and Mycology Program, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile.,Department of Pediatrics and Pediatric Surgery, Dr. Luis Calvo Mackenna Hospital, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Roberto Vidal
- Microbiology and Mycology Program, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago, Chile.,ANID - Millennium Science Initiative Program - Millennium Nucleus in the Biology of Intestinal Microbiota, Santiago, Chile
| | - Miguel O'Ryan
- Host-Pathogen Interaction Laboratory, Microbiology and Mycology Program, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
12
|
Contribution of Gut Microbiota to Immune Tolerance in Infants. J Immunol Res 2022; 2021:7823316. [PMID: 34993254 PMCID: PMC8727111 DOI: 10.1155/2021/7823316] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 11/18/2021] [Indexed: 02/07/2023] Open
Abstract
The prevalence of food allergy has increased in recent years, especially among the pediatric population. Differences in the gut microbiota composition between children with FA and healthy children have brought this topic into the spotlight as a possible explanation for the increase in FA. The gut microbiota characteristics are acquired through environmental interactions starting early in life, such as type of delivery during birth and breastfeeding. The microbiota features may be shaped by a plethora of immunomodulatory mechanisms, including a predominant role of Tregs and the transcription factor FOXP3. Additionally, a pivotal role has been given to vitamin A and butyrate, the main anti-inflammatory metabolite. These observations have led to the study and development of therapies oriented to modifying the microbiota and metabolite profiles, such as the use of pre- and probiotics and the determination of their capacity to induce tolerance to allergens that are relevant to FA. To date, evidence supporting these approaches in humans is scarce and inconclusive. Larger cohorts and dose-titration studies are mandatory to evaluate whether the observed changes in gut microbiota composition reflect medical recovery and increased tolerance in pediatric patients with FA. In this article, we discuss the establishment of the microbiota, the immunological mechanisms that regulate the microbiota of children with food allergies, and the evidence in research focused on its regulation as a means to achieve tolerance to food allergens.
Collapse
|
13
|
Laursen MF. Gut Microbiota Development: Influence of Diet from Infancy to Toddlerhood. ANNALS OF NUTRITION & METABOLISM 2021; 77:1-14. [PMID: 34461613 DOI: 10.1159/000517912] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/15/2021] [Indexed: 11/19/2022]
Abstract
Early life is a critical period as our gut microbiota establishes here and may impact both current and future health. Thus, it is of importance to understand how different factors govern the complex microbial colonization patterns in this period. The gut microbiota changes substantially during infancy and toddlerhood in terms of both taxonomic composition and diversity. This developmental trajectory differs by a variety of factors, including term of birth, mode of birth, intake of antibiotics, presence of furred pets, siblings and family members, host genetics, local environment, geographical location, and maternal and infant/toddler diet. The type of milk feeding and complementary feeding is particularly important in early and late infancy/toddlerhood, respectively. Breastfeeding, due to the supply of human milk oligosaccharide into the gut, promotes the growth of specific human milk oligosaccharide (HMO)-utilizing Bifidobacterium species that dominate the ecosystem as long as the infant is primarily breastfed. These species perform saccharolytic fermentation in the gut and produce metabolites with physiological effects that may contribute to protection against infectious and immune-related diseases. Formula feeding, due to its lack of HMOs and higher protein content, give rise to a more diverse gut microbiota that contains more opportunistic pathogens and results in a more proteolytic metabolism in the gut. Complementary feeding, due to the introduction of dietary fibers and new protein sources, induces a shift in the gut microbiota and metabolism away from the milk-adapted and toward a more mature and diverse adult-like community with increased abundances of short chain fatty acid-producing bacterial taxa. While the physiological implication of these complementary diet-induced changes remains to be established, a few recent studies indicate that an inadequately matured gut microbiota may be causally related to poor growth and development. Further studies are required to expand our knowledge on interactions between diet, gut microbiota, and health in the early life setting.
Collapse
|
14
|
Yang K, He S, Dong W. Gut microbiota and bronchopulmonary dysplasia. Pediatr Pulmonol 2021; 56:2460-2470. [PMID: 34077996 DOI: 10.1002/ppul.25508] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/02/2021] [Accepted: 05/16/2021] [Indexed: 12/20/2022]
Abstract
Bronchopulmonary dysplasia is a relatively common and severe complication of prematurity, and its pathogenesis remains ambiguous. Revolutionary advances in microbiological analysis techniques, together with the growing sophistication of the gut-lung axis hypothesis, have resulted in more studies linking gut microbiota dysbiosis to the occurrence and development of bronchopulmonary dysplasia. The present article builds on current findings to examine the intrinsic associations between gut microbiota and bronchopulmonary dysplasia. Gut microbiota dysbiosis may insult the intestinal barrier, triggering inflammation, metabolic disturbances, and malnutrition, consequences of which might impact bronchopulmonary dysplasia by altering the gut-lung axis. By evaluating the potential mechanisms, new therapeutic targets and potential therapeutic modalities for bronchopulmonary dysplasia can be identified from a microecological perspective.
Collapse
Affiliation(s)
- Kun Yang
- Department of Pediatrics, Division of Neonatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Shasha He
- Department of Pediatrics, Division of Neonatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wenbin Dong
- Department of Pediatrics, Division of Neonatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
15
|
Strobel KM, Romero T, Kramer K, Fernandez E, Rottkamp C, Uy C, Keller R, Moyer L, Poulain F, Kim JH, DeUgarte DA, Calkins KL. Growth Failure Prevalence in Neonates with Gastroschisis : A Statewide Cohort Study. J Pediatr 2021; 233:112-118.e3. [PMID: 33647253 PMCID: PMC8154735 DOI: 10.1016/j.jpeds.2021.02.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVES To perform a multicenter study to assess growth failure in hospitalized infants with gastroschisis. STUDY DESIGN This study included neonates with gastroschisis within sites in the University of California Fetal Consortium. The study's primary outcome was growth failure at hospital discharge, defined as a weight or length z score decrease >0.8 from birth. Regression analysis was performed to assess changes in z scores over time. RESULTS Among 125 infants with gastroschisis, the median gestational age was 37 weeks (IQR 35-37). Length of stay was 32 days (23-60); 55% developed weight or length growth failure at discharge (28% had weight growth failure, 42% had length growth failure, and 15% had both weight and length growth failure). Weight and length z scores at 14 days, 30 days, and discharge were less than birth (P < .01 for all). Weight and length z scores declined from birth to 30 days (-0.10 and -0.11 z score units/week, respectively, P < .001). Length growth failure at discharge was associated with weight and length z score changes over time (P < .05 for both). Lower gestational age was associated with weight growth failure (OR 0.70 for each gestational age week, 95% CI 0.55-0.89, P = .004). CONCLUSIONS Growth failure, in particular linear growth failure, is common in infants with gastroschisis. These data suggest the need to improve nutritional management in these infants.
Collapse
Affiliation(s)
- Katie M Strobel
- Division of Neonatology and Developmental Biology, Department of Pediatrics, University of California Los Angeles, Los Angeles, CA.
| | - Tahmineh Romero
- Division of General Internal Medicine and Health Services Research, Department of Medicine Statistics Core, University of California Los Angeles, Los Angeles, CA
| | - Katelin Kramer
- Division of Neonatology, Department of Pediatrics, University of California San Francisco, San Francisco, CA
| | - Erika Fernandez
- Division of Neonatology, Department of Pediatrics, University of California San Diego, San Diego, CA
| | - Catherine Rottkamp
- Division of Neonatology, Department of Pediatrics, University of California Davis, Davis, CA
| | - Cherry Uy
- Division of Neonatal/Perinatal Medicine, Department of Pediatrics, University of California Irvine, Irvine, CA
| | - Roberta Keller
- Division of Neonatology, Department of Pediatrics, University of California San Francisco, San Francisco, CA
| | - Laurel Moyer
- Division of Neonatology, Rady Children's Hospital, San Diego, CA
| | - Francis Poulain
- Division of Neonatology, Department of Pediatrics, University of California Davis, Davis, CA
| | - Jae H Kim
- Division of Neonatology, Department of Pediatrics, University of California San Diego, San Diego, CA
| | - Daniel A DeUgarte
- Division of Pediatric Surgery, Department of Surgery, University of California Los Angeles, Los Angeles, CA
| | - Kara L Calkins
- Division of Neonatology and Developmental Biology, Department of Pediatrics, University of California Los Angeles, Los Angeles, CA
| |
Collapse
|
16
|
Atyeo C, Alter G. The multifaceted roles of breast milk antibodies. Cell 2021; 184:1486-1499. [PMID: 33740451 DOI: 10.1016/j.cell.2021.02.031] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/07/2021] [Accepted: 02/12/2021] [Indexed: 12/20/2022]
Abstract
Neonates are born with an immature immune system and rely on the transfer of immunity from their mothers. Maternal antibodies are transferred via the placenta and breast milk. Although the role of placentally transferred immunoglobulin G (IgG) is established, less is known about the selection of antibodies transferred via breast milk and the mechanisms by which they provide protection against neonatal disease. Evidence suggests that breast milk antibodies play multifaceted roles, preventing infection and supporting the selection of commensals and tolerizing immunity during infancy. Here, we discuss emerging data related to the importance of breast milk antibodies in neonatal immunity and development.
Collapse
Affiliation(s)
- Caroline Atyeo
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; PhD Program in Virology, Division of Medical Sciences, Harvard University, Boston, MA, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.
| |
Collapse
|
17
|
Human Milk-Fed Piglets Have a Distinct Small Intestine and Circulatory Metabolome Profile Relative to That of Milk Formula-Fed Piglets. mSystems 2021; 6:6/1/e01376-20. [PMID: 33563783 PMCID: PMC7883546 DOI: 10.1128/msystems.01376-20] [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] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Exclusive HM feeding for newborns is recommended at least for the first 6 months of life. However, when breastfeeding is not possible, MF is recommended as a substitute. The impact of human milk (HM) feeding compared with cow’s milk formula (MF) feeding on small intestinal and circulatory metabolome patterns has not been fully investigated. Therefore, 2-day-old male piglets were fed HM or MF (n = 26/group) from postnatal day 2 (PND 2) through 21 and were weaned to a solid diet until PND 51. The small intestine (gastrointestinal [GI]) contents, serum, and urine were collected from subsets of piglets at PND 21 and PND 51. Samples were subjected to primary metabolomics analyses at the West Coast Metabolomics Center, UC Davis. The metabolome data assessment and the statistical analyses were performed with MetaboAnalyst software. Compared with MF feeding, at PND 21, HM feeding resulted in a higher abundance of fucose in the jejunum and urine and a greater concentration of myo-inositol in serum. In HM-fed piglets, 1,5-anhydroglucitol was higher in the duodenum, serum, and urine at PND 21. Additionally, the HM group had higher levels of urinary kynurenic acid at PND 21. Correlations between bacterial genera and altered metabolites in ileum revealed that Turicibacter sp. and Campylobacter sp. were positively correlated with maltotriose and panose at PND 21, while ileal Campylobacter sp. was negatively correlated with fumaric acid. At PND 51, no significant metabolites were identified between HM and MF diet groups. The metabolites associated with the neonatal diets may serve as the substrates and signals that contribute to the physiological effects in HM and MF during infancy, with a subset reflecting diet-associated differences in microbial metabolism and ecology. IMPORTANCE Exclusive HM feeding for newborns is recommended at least for the first 6 months of life. However, when breastfeeding is not possible, MF is recommended as a substitute. Due to the challenges associated with sample collection from infants fed HM or MF, their gut metabolism is poorly understood. Thus, an established piglet model from our team was used to determine the metabolite profile in relation to host, diet, and microbiota. The current study is the first to provide novel insights across the small intestine metabolism and its association with circulatory metabolites in the HM group relative to the MF group at the weaning and postweaning period. Data also demonstrate that during the neonatal period, diet, host, and microbial metabolism contribute to the lumen and circulatory metabolite profile. Furthermore, small intestinal lumen metabolome can be tracked in the urine as a biomarker of dietary differences, which would be a useful tool for clinical interventions.
Collapse
|
18
|
Rosa F, Matazel KS, Bowlin AK, Williams KD, Elolimy AA, Adams SH, Bode L, Yeruva L. Neonatal Diet Impacts the Large Intestine Luminal Metabolome at Weaning and Post-Weaning in Piglets Fed Formula or Human Milk. Front Immunol 2020; 11:607609. [PMID: 33365033 PMCID: PMC7750455 DOI: 10.3389/fimmu.2020.607609] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 11/05/2020] [Indexed: 12/13/2022] Open
Abstract
The impact of human milk (HM) or dairy milk-based formula (MF) on the large intestine’s metabolome was not investigated. Two-day old male piglets were randomly assigned to HM or MF diet (n = 26/group), from postnatal day (PND) 2 through 21 and weaned to a solid diet until PND 51. Piglets were euthanized at PND 21 and PND 51, luminal contents of the cecum, proximal (PC) and distal colons (DC), and rectum were collected and subjected to metabolomics analysis. Data analyses were performed using Metaboanalyst. In comparison to MF, the HM diet resulted in higher levels of fatty acids in the lumen of the cecum, PC, DC, and rectum at PND 21. Glutamic acid was greater in the lumen of cecum, PC, and DC relative to the MF group at PND 21. Also, spermidine was higher in the DC and rectal contents of HM relative to MF at PND 21. MF diet resulted in greater abundances of amino acids in the cecal lumen relative to HM diet at PND 21. Additionally, several sugar metabolites were higher in various regions of the distal gut of MF fed piglets relative to HM group at PND 21. In contrast, at PND 51, in various regions there were higher levels of erythritol, maltotriose, isomaltose in HM versus MF fed piglets. This suggests a post weaning shift in sugar metabolism that is impacted by neonatal diet. The data also suggest that infant diet type and host-microbiota interactions likely influence the lower gut metabolome.
Collapse
Affiliation(s)
- Fernanda Rosa
- Arkansas Children's Nutrition Center, Little Rock, AR, United States.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Katelin S Matazel
- Arkansas Children's Nutrition Center, Little Rock, AR, United States.,Arkansas Children's Research Institute, Little Rock, AR, United States
| | - Anne K Bowlin
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Keith D Williams
- Arkansas Children's Nutrition Center, Little Rock, AR, United States.,Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock AR, United States
| | - Ahmed A Elolimy
- Arkansas Children's Nutrition Center, Little Rock, AR, United States.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Sean H Adams
- Arkansas Children's Nutrition Center, Little Rock, AR, United States.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Lars Bode
- Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence, University of California San Diego, La Jolla, CA, United States.,Department of Pediatrics, University of California San Diego, La Jolla, CA, United States
| | - Laxmi Yeruva
- Arkansas Children's Nutrition Center, Little Rock, AR, United States.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Arkansas Children's Research Institute, Little Rock, AR, United States
| |
Collapse
|
19
|
Verhaeghe R, George K, Westerman M, Olbina G, McCann D, Parrow N, Pincus E, Havranek T, Fleming RE. Hepcidin Status at 2 Months in Infants Fed Breast Milk Compared with Formula. Neonatology 2020; 117:474-479. [PMID: 32818935 DOI: 10.1159/000508447] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/05/2020] [Indexed: 11/19/2022]
Abstract
INTRODUCTION The basis for the superior absorption of iron from breast milk compared with infant formulas is unclear. The hormone hepcidin downregulates dietary iron absorption. Hepcidin production increases with increased body iron status (reflected in serum ferritin levels). We hypothesized that serum hepcidin levels are suppressed relative to iron status in infants fed breast milk compared with formula. METHODS Subjects were healthy infants presenting for routine 2-month clinic visit and strictly fed either breast milk or standard infant formula. Urinary hepcidin and ferritin levels (reflective of serum levels) were analyzed and compared across the breast milk- and formula-fed groups. The relationship between urinary hepcidin and ferritin levels within each group was analyzed by linear regression. RESULTS Twenty-four subjects were enrolled in each group. The median urinary hepcidin level in the group fed breast milk was lower than in formula (130 vs. 359 ng hepcidin/mg creatinine, p < 0.05). However, the median ferritin levels were similar (2.1 vs. 1.9 ng/mL). Within each group, urinary hepcidin correlated with urinary ferritin (r = 0.5, p < 0.05 for each group); however, the slope of the regression line was lower in the group fed breast milk compared with formula (p < 0.005). CONCLUSION Despite similar urinary ferritin levels, urinary hepcidin levels are lower at 2 months in infants fed breast milk compared with infants fed formula. Hepcidin levels correlate with iron status in each group; however, this relationship is relatively dampened in infants fed breast milk. We speculate that relatively lower infant hepcidin contributes to the superior efficiency of iron absorption from breast milk.
Collapse
Affiliation(s)
- Rebecca Verhaeghe
- Pediatrics/Neonatology, Saint Louis University, St. Louis, Missouri, USA
| | - Kandie George
- Pediatrics/Neonatology, Albert Einstein University/Montefiore, Bronx, New York, USA
| | | | | | - Diane McCann
- Intrinsic LifeSciences, La Jolla, California, USA
| | - Nermi Parrow
- Pediatrics/Neonatology, Saint Louis University, St. Louis, Missouri, USA
| | - Elisa Pincus
- Pediatrics/Neonatology, Saint Louis University, St. Louis, Missouri, USA
| | - Tomas Havranek
- Pediatrics/Neonatology, Albert Einstein University/Montefiore, Bronx, New York, USA
| | - Robert E Fleming
- Pediatrics/Neonatology, Saint Louis University, St. Louis, Missouri, USA,
| |
Collapse
|
20
|
Elolimy AA, Washam C, Byrum S, Chen C, Dawson H, Bowlin AK, Randolph CE, Saraf MK, Yeruva L. Formula Diet Alters the Ileal Metagenome and Transcriptome at Weaning and during the Postweaning Period in a Porcine Model. mSystems 2020; 5:e00457-20. [PMID: 32753508 PMCID: PMC7406227 DOI: 10.1128/msystems.00457-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/21/2020] [Indexed: 01/05/2023] Open
Abstract
Exclusive breastfeeding impacts the intestinal microbiome and is associated with a better immune function than is seen with milk formula (MF) feeding in infants and yet with mechanisms poorly defined. The porcine model was used to evaluate the impact of MF on ileum microbial communities and gene expression relative to human milk (HM)-fed piglets. Fifty-two Dutch Landrace male piglets were fed an isocaloric diet of either HM (n = 26) or MF (n = 26) from day 2 through day 21 of age and weaned to a solid diet until day 51. Eleven piglets from each group were euthanized at day 21, while the remaining piglets (HM, n = 15; MF, n = 15) were euthanized at day 51 to collect ileal epithelium (EP) scrapings and ileal (IL) tissues. The epithelial mucosa was subjected to shotgun metagenome sequencing, and EP and IL tissues were used for transcriptome analysis. On day 21, transcriptome data revealed that the levels of pathways involved in inflammation and apoptosis were significantly higher in MF piglets than in HM piglets, whereas the levels of tight junctions and pathogen detection systems were lower in MF piglets than in HM piglets. The MF impacts on the small intestine were maintained over the postweaning period (day 51) as indicated by higher levels of Dialister invisus bacteria and higher levels of expression of genes associated with inflammation and apoptosis pathways relative to HM group. The current study demonstrated that MF might impact local intestinal inflammation, apoptosis, and tight junctions and might suppress pathogen recognition in the small intestine compared with HM.IMPORTANCE Exclusive human milk (HM) breastfeeding for the first 6 months of age in infants is recommended to improve health outcomes during early life and beyond. When women are unable to provide sufficient HM, milk formula (MF) is often recommended as a complementary or alternative source of nutrition. Previous studies in piglets demonstrated that MF alters the gut microbiome and induces inflammatory cytokine production. The links between MF feeding, gut microbiome, and inflammation status are unclear due to challenges associated with the collection of intestinal samples from human infants. The current report provides the first insight into MF-microbiome-inflammation connections in the small intestine compared with HM feeding using a porcine model. The present results showed that, compared with HM, MF might impact immune function through the induction of ileal inflammation, apoptosis, and tight junction disruptions and likely compromised immune defense against pathogen detection in the small intestine relative to piglets that were fed HM.
Collapse
Affiliation(s)
- Ahmed A Elolimy
- Arkansas Children's Nutrition Center, Little Rock, Arkansas, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Charity Washam
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Stephanie Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Celine Chen
- Diet, Genomics & Immunology Laboratory, USDA-ARS Beltsville Human Nutrition Research Center, Beltsville, Maryland, USA
| | - Harry Dawson
- Diet, Genomics & Immunology Laboratory, USDA-ARS Beltsville Human Nutrition Research Center, Beltsville, Maryland, USA
| | - Anne K Bowlin
- Arkansas Children's Nutrition Center, Little Rock, Arkansas, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | | | - Manish K Saraf
- Arkansas Children's Nutrition Center, Little Rock, Arkansas, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Laxmi Yeruva
- Arkansas Children's Nutrition Center, Little Rock, Arkansas, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Arkansas Children's Research Institute, Little Rock, Arkansas, USA
| |
Collapse
|
21
|
Machate DJ, Figueiredo PS, Marcelino G, Guimarães RDCA, Hiane PA, Bogo D, Pinheiro VAZ, de Oliveira LCS, Pott A. Fatty Acid Diets: Regulation of Gut Microbiota Composition and Obesity and Its Related Metabolic Dysbiosis. Int J Mol Sci 2020; 21:E4093. [PMID: 32521778 PMCID: PMC7312778 DOI: 10.3390/ijms21114093] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 03/27/2020] [Accepted: 03/27/2020] [Indexed: 02/06/2023] Open
Abstract
Long-term high-fat dietary intake plays a crucial role in the composition of gut microbiota in animal models and human subjects, which affect directly short-chain fatty acid (SCFA) production and host health. This review aims to highlight the interplay of fatty acid (FA) intake and gut microbiota composition and its interaction with hosts in health promotion and obesity prevention and its related metabolic dysbiosis. The abundance of the Bacteroidetes/Firmicutes ratio, as Actinobacteria and Proteobacteria species are associated with increased SCFA production, reported high-fat diet rich in medium-chain fatty acids (MCFAs), monounsaturated fatty acids (MUFAs), and n-3 polyunsaturated fatty acids (PUFAs) as well as low-fat diets rich in long-chain fatty acids (LCFAs). SCFAs play a key role in health promotion and prevention and, reduction and reversion of metabolic syndromes in the host. Furthermore, in this review, we discussed the type of fatty acids and their amount, including the administration time and their interplay with gut microbiota and its results about health or several metabolic dysbioses undergone by hosts.
Collapse
Affiliation(s)
- David Johane Machate
- Graduate Program in Biotechnology and Biodiversity in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul, Campo Grande 79079-900, Brazil; (D.J.M.); (A.P.)
| | - Priscila Silva Figueiredo
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul, Campo Grande 79079-900, Brazil; (P.S.F.); (G.M.); (P.A.H.); (D.B.); (V.A.Z.P.)
| | - Gabriela Marcelino
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul, Campo Grande 79079-900, Brazil; (P.S.F.); (G.M.); (P.A.H.); (D.B.); (V.A.Z.P.)
| | - Rita de Cássia Avellaneda Guimarães
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul, Campo Grande 79079-900, Brazil; (P.S.F.); (G.M.); (P.A.H.); (D.B.); (V.A.Z.P.)
| | - Priscila Aiko Hiane
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul, Campo Grande 79079-900, Brazil; (P.S.F.); (G.M.); (P.A.H.); (D.B.); (V.A.Z.P.)
| | - Danielle Bogo
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul, Campo Grande 79079-900, Brazil; (P.S.F.); (G.M.); (P.A.H.); (D.B.); (V.A.Z.P.)
| | - Verônica Assalin Zorgetto Pinheiro
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul, Campo Grande 79079-900, Brazil; (P.S.F.); (G.M.); (P.A.H.); (D.B.); (V.A.Z.P.)
| | | | - Arnildo Pott
- Graduate Program in Biotechnology and Biodiversity in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul, Campo Grande 79079-900, Brazil; (D.J.M.); (A.P.)
| |
Collapse
|
22
|
Parker LA, Weaver M, Torrazza RJM, Shuster J, Li N, Krueger C, Neu J. Effect of Aspiration and Evaluation of Gastric Residuals on Intestinal Inflammation, Bleeding, and Gastrointestinal Peptide Level. J Pediatr 2020; 217:165-171.e2. [PMID: 31757473 PMCID: PMC7012706 DOI: 10.1016/j.jpeds.2019.10.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 09/01/2019] [Accepted: 10/11/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To determine the effect of gastric residual aspiration and evaluation on preterm very low birth weight infants' gastrointestinal function, intestinal inflammation, and gastrointestinal mucosal bleeding. STUDY DESIGN This single-center, randomized trial compared omission of gastric residuals vs prefeed gastric residuals in 143 infants ≤32 weeks of gestation with a birthweight of ≤1250 g for 6 weeks after birth. Serum levels of gastrin and motilin were collected between 14 and 21 days of life. Stools were collected at 3 and 6 weeks of age and analyzed for calprotectin and S100A12 levels. All stools were tested for occult blood for 6 weeks. RESULTS Means for gastrin (P = .999) and motilin (P = .694) were similar between groups and there were no statistically significant differences in adjusted means for transformed calprotectin (P = .580), and S100A12 (P = .212). Both calprotectin (P = .003) and S100A12 (P = .002) increased from week 3 to week 6. The mean percentage of stools positive for occult blood (P = .888) were similar between the groups. CONCLUSIONS Gastrointestinal function, intestinal inflammation, and gastrointestinal mucosal bleeding were similar whether aspiration and evaluation of gastric residuals were eliminated or not, suggesting routinely evaluating gastric residuals before every feeding may be unnecessary. TRIAL REGISTRATION CLINICALTRIALS.GOV:: NCT01863043.
Collapse
Affiliation(s)
- Leslie A. Parker
- College of Nursing, University of Florida; No conflict of interest
| | - Michael Weaver
- College of Nursing, University of Florida; No conflict of interest
| | - Roberto J. Murgas Torrazza
- Sistema Nacional de Investigacion de Panama (SNI). Secretaria Nacional de Ciencia Tecnologia e Innovacion (SENACYT); No conflict of interest
| | - Jonathon Shuster
- Department of Health Outcomes and Bioinformatics, University of Florida; No conflict of interest
| | - Nan Li
- Department of Pediatrics, University of Florida; No conflict of interest
| | - Charlene Krueger
- College of Nursing, University of Florida; No conflict of interest
| | - Josef Neu
- Department of Pediatrics, University of Florida; Conflict: Research grant from Infant Bacterial Therapeutics
| |
Collapse
|
23
|
Huang X, Zhu B, Jiang T, Yang C, Qiao W, Hou J, Han Y, Xiao H, Chen L. Improved Simple Sample Pretreatment Method for Quantitation of Major Human Milk Oligosaccharides Using Ultrahigh Pressure Liquid Chromatography with Fluorescence Detection. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:12237-12244. [PMID: 31560847 DOI: 10.1021/acs.jafc.9b03445] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Human milk oligosaccharides (HMOs) maintain and promote infant health. Most of the current methods for HMOs quantitation require labor-intensive and time-consuming steps for sample preparation. This study presents two very simple and easy-to-operate pretreatment methods, requiring either ultrafiltration or centrifugation to separate free oligosaccharides from whole fat human milk and other milk matrix before oligosaccharides labeling for quantifying HMOs using ultrahigh pressure liquid chromatography with fluorescence detection. A single chromatography run quantified 15 sialylated and neutral HMOs with high sensitivity (with an LOD less than 8 pg for all HMOs tested: about 1 pg for 2'-fucosyllactose, 3-fucosyllactose, 4'-galactosyllactose, 3'-galactosyllactose, and 6'-galactosyllactose) and good linearity with coefficient of correlation above 0.999. Accuracy and precision were satisfactory for both pretreatment methods. Overall, the centrifugation pretreatment was efficient and reliable for samples with high levels of oligosaccharides, and the ultrafiltration pretreatment was especially suitable for samples with low oligosaccharide abundance.
Collapse
Affiliation(s)
- Xunwen Huang
- National Engineering Center of Dairy for Maternal and Child Health , Beijing Sanyuan Foods Co. Ltd. , Beijing 100163 , P. R. China
- Beijing Engineering Research Center of Dairy , Beijing Sanyuan Foods Co. Ltd. , Beijing 100163 , P. R. China
- Institute of Microbiology , China Academy of Sciences , Beijing 100101 , P. R. China
| | - Baoli Zhu
- Institute of Microbiology , China Academy of Sciences , Beijing 100101 , P. R. China
| | - Tiemin Jiang
- National Engineering Center of Dairy for Maternal and Child Health , Beijing Sanyuan Foods Co. Ltd. , Beijing 100163 , P. R. China
- Beijing Engineering Research Center of Dairy , Beijing Sanyuan Foods Co. Ltd. , Beijing 100163 , P. R. China
| | - Chunying Yang
- National Engineering Center of Dairy for Maternal and Child Health , Beijing Sanyuan Foods Co. Ltd. , Beijing 100163 , P. R. China
- Beijing Engineering Research Center of Dairy , Beijing Sanyuan Foods Co. Ltd. , Beijing 100163 , P. R. China
| | - Weicang Qiao
- National Engineering Center of Dairy for Maternal and Child Health , Beijing Sanyuan Foods Co. Ltd. , Beijing 100163 , P. R. China
- Beijing Engineering Research Center of Dairy , Beijing Sanyuan Foods Co. Ltd. , Beijing 100163 , P. R. China
| | - Juncai Hou
- College of Food Science , Northeast Agricultural University , Harbin 150030 , P. R. China
| | - Yanhui Han
- Department of Food Science , University of Massachusetts , Amherst , Massachusetts 010003 , United States
| | - Hang Xiao
- Department of Food Science , University of Massachusetts , Amherst , Massachusetts 010003 , United States
| | - Lijun Chen
- National Engineering Center of Dairy for Maternal and Child Health , Beijing Sanyuan Foods Co. Ltd. , Beijing 100163 , P. R. China
- Beijing Engineering Research Center of Dairy , Beijing Sanyuan Foods Co. Ltd. , Beijing 100163 , P. R. China
| |
Collapse
|