1
|
Swaney MH, Steidl OR, Tackett A, Fye S, Lee KE, Ong IM, Bendixsen C, Spicer G, DeLine J, Gern JE, Lucey J, Seroogy CM, Kalan L, Singh AM. Farm exposure is associated with human breast milk immune profile and microbiome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.14.618271. [PMID: 39464075 PMCID: PMC11507701 DOI: 10.1101/2024.10.14.618271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Prenatal and early life farm exposure, and breastfeeding, are associated with protection from allergic diseases. We hypothesize that farm exposure influences the human breast milk microbiome and immune proteins. The immune protein profiles and microbial communities of 152 human breast milk samples were compared among three maternal farm exposure groups (traditional agrarian, farm, and non-farm) in rural Wisconsin to identify signatures associated with farm status and atopic disease. We found significant differences between farm groups for 23 immune proteins (p-adj<0.05), microbiome diversity (p=2.2E-05), and microbiome richness (p=8.0e-06). Traditional agrarian human breast milk had the highest immune protein levels and microbiome diversity and richness, followed by farm and non-farm human breast milk. Furthermore, Gram-positive bacterial species correlated with IL-23 mediated signaling events (p-adj<1.0E-05). These data suggest that increased farm exposures promotes human breast milk that is more microbially-diverse and rich in immune-associated proteins, ultimately influencing immune development in the infant.
Collapse
|
2
|
Angelopoulou A, Harris HMB, Warda AK, O'Shea CA, Lavelle A, Ryan CA, Dempsey E, Stanton C, Hill C, Ross RP. Somatic cell count as an indicator of subclinical mastitis and increased inflammatory response in asymptomatic lactating women. Microbiol Spectr 2024; 12:e0405123. [PMID: 39189754 PMCID: PMC11448179 DOI: 10.1128/spectrum.04051-23] [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: 12/04/2023] [Accepted: 07/02/2024] [Indexed: 08/28/2024] Open
Abstract
Subclinical mastitis is an asymptomatic inflammatory condition that can be difficult to define and diagnose. In the dairy industry, subclinical mastitis is diagnosed by milk somatic cell counts (SCCs) of ≥250,000 cells mL-1. In this pilot study, we assessed the efficacy of this index to identify human subclinical mastitis by comparing SCC levels with the inflammatory response [interleukin-8 (IL-8) levels] in 37 samples from asymptomatic and 10 clinical mastitis (CM) lactating women. The milk microbiota was determined by 16S rRNA gene sequencing. The SCC of CM samples ranged from 310,000 to 6,600,000 cells mL-1. However, 14 of 37 (37.8%) asymptomatic samples had high SCC (250,000-460,000 cells mL-1), indicating subclinical mastitis. SCC levels significantly (P < 0.001) and positively correlated with milk IL-8 levels reflecting the escalating inflammatory response across subclinical and clinical mastitis samples. Samples with an SCC of ≥250,000 cells mL-1 showed significant increases in IL-8 responses when compared with milk samples from healthy women. The milk microbiome of CM samples was dominated by streptococcal and staphylococcal species (89.9% combined median relative abundance). In contrast, the combined median streptococcal/staphylococcal relative levels were 75.4% and 66.3% in milks from asymptomatic (subclinical mastitis) and healthy groups, respectively. The Streptococcus genus was increased in samples with an SCC of ≥250,000, although this should be interpreted with caution. Thus, the index of ≥250,000 somatic cells mL-1 could be a reliable indicator of subclinical mastitis in humans and should aid future studies investigating the impact of subclinical mastitis on maternal health, breastfeeding behaviors, infant health, and development. IMPORTANCE This pilot study suggests that SCC at a level of (greater than or equal to) 250,000 cells mL-1, as used in the dairy industry, is a suitable index to identify asymptomatic subclinical mastitis in lactating women since it reflects a significant increase in the inflammatory response compared to milk samples from healthy women. Using this index should aid studies into the short- and long-term consequences of subclinical mastitis for mother and infant.
Collapse
Affiliation(s)
- Angeliki Angelopoulou
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Hugh M B Harris
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Alicja K Warda
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Carol-Anne O'Shea
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Neonatology, Cork University Maternity Hospital, Cork, Ireland
| | - Aonghus Lavelle
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - C Anthony Ryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Neonatology, Cork University Maternity Hospital, Cork, Ireland
| | - Eugene Dempsey
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Neonatology, Cork University Maternity Hospital, Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Food Biosciences, Teagasc Food Research Centre, Fermoy, Co Cork, Ireland
| | - Colin Hill
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - R Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| |
Collapse
|
3
|
Ostojic SM. Establishing Reference Intakes for Creatine in Infants Aged 0 to 12 Months. Nutr Rev 2024:nuae124. [PMID: 39271173 DOI: 10.1093/nutrit/nuae124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024] Open
Abstract
Creatine is recognized as a conditionally essential nutrient in certain populations; however, there is a lack of established reference values across different life stages. Infants rely exclusively on dietary creatine from human milk for their first 6 months; evaluating creatine adequacy in this population can be estimated based on preliminary data regarding the intake needed to promote optimal growth. This special article explores creatine requirements for infants aged 0 to 12 months, presents a summary of creatine content in human milk, and proposes reference intakes for creatine in this population.
Collapse
Affiliation(s)
- Sergej M Ostojic
- Department of Nutrition and Public Health, University of Agder, 4630 Kristiansand, Norway
- Applied Bioenergetics Lab, Faculty of Sport and PE, University of Novi Sad, 21000 Novi Sad, Serbia
- Faculty of Health Sciences, University of Pécs, 7622 Pécs, Hungary
| |
Collapse
|
4
|
Konieczna M, Koryszewska-Bagińska A, Bzikowska-Jura A, Chmielewska-Jeznach M, Jarzynka S, Olędzka G. Modifiable and Non-Modifiable Factors That Affect Human Milk Oligosaccharides Composition. Nutrients 2024; 16:2887. [PMID: 39275203 PMCID: PMC11397269 DOI: 10.3390/nu16172887] [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: 07/25/2024] [Revised: 08/22/2024] [Accepted: 08/27/2024] [Indexed: 09/16/2024] Open
Abstract
Human milk, the gold standard in infant nutrition, is a unique fluid that provides essential nutrients such as lactose, lipids, proteins, and free oligosaccharides. While its primary role is nutritional, it also protects against pathogens. This protection mainly comes from immunoglobulins, with human milk oligosaccharides (HMOs) providing additional support by inhibiting pathogen binding to host cell ligands. The prebiotic and immune-modulatory activity of HMOs strongly depends on their structure. Over 200 individual structures have been identified so far, with the composition varying significantly among women. The structure and composition of HMOs are influenced by factors such as the Lewis blood group, secretor status, and the duration of nursing. HMO profiles are heavily influenced by maternal phenotypes, which are defined based on the expression of two specific fucosyltransferases. However, recent data have shown that HMO content can be modified by various factors, both changeable and unchangeable, including diet, maternal age, gestational age, mode of delivery, breastfeeding frequency, and race. The first part of this overview presents the historical background of these sugars and the efforts by scientists to extract them using the latest chromatography methods. The second part is divided into subchapters that examine modifiable and non-modifiable factors, reviewing the most recent articles on HMO composition variations due to specific reasons and summarizing potential future challenges in conducting these types of studies.
Collapse
Affiliation(s)
- Małgorzata Konieczna
- Department of Medical Biology, Medical University of Warsaw, 00-575 Warsaw, Poland
| | | | - Agnieszka Bzikowska-Jura
- Department of Medical Biology, Medical University of Warsaw, 00-575 Warsaw, Poland
- Laboratory of Human Milk and Lactation Research, Department of Medical Biology, Medical University of Warsaw, 00-575 Warsaw, Poland
| | | | - Sylwia Jarzynka
- Department of Medical Biology, Medical University of Warsaw, 00-575 Warsaw, Poland
| | - Gabriela Olędzka
- Department of Medical Biology, Medical University of Warsaw, 00-575 Warsaw, Poland
| |
Collapse
|
5
|
Schulkers Escalante K, Bai-Tong SS, Allard SM, Ecklu-Mensah G, Sanchez C, Song SJ, Gilbert J, Bode L, Dorrestein P, Knight R, Gonzalez DJ, Leibel SA, Leibel SL. The impact of breastfeeding on the preterm infant's microbiome and metabolome: a pilot study. Pediatr Res 2024:10.1038/s41390-024-03440-9. [PMID: 39138352 DOI: 10.1038/s41390-024-03440-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 06/23/2024] [Accepted: 07/10/2024] [Indexed: 08/15/2024]
Abstract
BACKGROUND Human milk is unquestionably beneficial for preterm infants. We investigated how the transition from tube to oral/breastfeeding impacts the preterm infants' oral and gut microbiome and metabolome. METHODS We analyzed stool, saliva, and milk samples collected from a cohort of preterm infants enrolled in the MAP Study, a prospective observational trial. The microbiome and metabolome of the samples were analyzed from 4 longitudinal sample time points, 2 during tube feeds only and 2 after the initiation of oral/breastfeeding. RESULTS We enrolled 11 mother-infant dyads (gestational age = 27.9 (23.4-32.2)) and analyzed a total of 39 stool, 44 saliva, and 43 milk samples over 4 timepoints. In saliva samples, there was a shift towards increased Streptococcus and decreased Staphylococcus after oral feeding/breastfeeding initiation (p < 0.05). Milk sample metabolites were strongly influenced by the route of feeding and milk type (p < 0.05) and represented the pathways of Vitamin E metabolism, Vitamin B12 metabolism, and Tryptophan metabolism. CONCLUSION Our analysis demonstrated that the milk and preterm infant's saliva microbiome and metabolome changed over the course of the first four to 5 months of life, coinciding with the initiation of oral/breastfeeds. IMPACT The microbiome and metabolome is altered in the infant's saliva but not their stool, and in mother's milk when feeds are transitioned from tube to oral/breastfeeding. We assessed the relationship between the gut and oral microbiome/metabolome with the milk microbiome/metabolome over a longitudinal period of time in preterm babies. Metabolites that changed in the infants saliva after the initiation of oral feeds have the potential to be used as biomarkers for disease risk.
Collapse
Affiliation(s)
| | - Shiyu S Bai-Tong
- Department of Pediatrics, University of California San Diego, Rady Children's Hospital, La Jolla, CA, USA
| | - Sarah M Allard
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | | | - Concepcion Sanchez
- Department of Pharmacology and the Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Se Jin Song
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
| | - Jack Gilbert
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Department of Pediatrics and Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA, USA
| | - Lars Bode
- Department of Pediatrics, Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (MOMI CORE), and the Human Milk Institute (HMI), University of California San Diego, La Jolla, CA, USA
| | - Pieter Dorrestein
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Collaborative Mass Spectrometry Innovation Center, University of California San Diego, La Jolla, CA, USA
| | - Rob Knight
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Collaborative Mass Spectrometry Innovation Center, University of California San Diego, La Jolla, CA, USA
| | - David J Gonzalez
- Department of Pharmacology and the Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Sydney A Leibel
- Department of Pediatrics, University of California San Diego, Rady Children's Hospital, La Jolla, CA, USA
| | - Sandra L Leibel
- Department of Pediatrics, University of California San Diego, Rady Children's Hospital, La Jolla, CA, USA.
| |
Collapse
|
6
|
Cohen A, Turjeman S, Levin R, Tal S, Koren O. Comparison of canine colostrum and milk using a multi-omics approach. Anim Microbiome 2024; 6:19. [PMID: 38650014 PMCID: PMC11034113 DOI: 10.1186/s42523-024-00309-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 04/09/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND A mother's milk is considered the gold standard of nutrition in neonates and is a source of cytokines, immunoglobulins, growth factors, and other important components, yet little is known about the components of canine milk, specifically colostrum, and the knowledge related to its microbial and metabolic profiles is particularly underwhelming. In this study, we characterized canine colostrum and milk microbiota and metabolome for several breeds of dogs and examined profile shifts as milk matures in the first 8 days post-whelping. RESULTS Through untargeted metabolomics, we identified 63 named metabolites that were significantly differentially abundant between days 1 and 8 of lactation. Surprisingly, the microbial compositions of the colostrum and milk, characterized using 16S rRNA gene sequencing, were largely similar, with only two differentiating genera. The shifts observed, mainly increases in several sugars and amino sugars over time and shifts in amino acid metabolites, align with shifts observed in human milk samples and track with puppy development. CONCLUSION Like human milk, canine milk composition is dynamic, and shifts are well correlated with developing puppies' needs. Such a study of the metabolic profile of canine milk, and its relation to the microbial community, provides insights into the changing needs of the neonate, as well as the ideal nutrition profile for optimal functionality. This information will add to the existing knowledge base of canine milk composition with the prospect of creating a quality, tailored milk substitute or supplement for puppies.
Collapse
Affiliation(s)
- Alisa Cohen
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Sondra Turjeman
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Rachel Levin
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Smadar Tal
- Koret School of Veterinary Medicine, The Hebrew University Veterinary Teaching Hospital, Hebrew University of Jerusalem, Rehovot, Israel
- Tel-Hai Academic College, Upper Galilee, Israel
| | - Omry Koren
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel.
- Kyung Hee University, Seoul, the Republic of Korea.
| |
Collapse
|
7
|
Chakraborty N. Metabolites: a converging node of host and microbe to explain meta-organism. Front Microbiol 2024; 15:1337368. [PMID: 38505556 PMCID: PMC10949987 DOI: 10.3389/fmicb.2024.1337368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 02/13/2024] [Indexed: 03/21/2024] Open
Abstract
Meta-organisms encompassing the host and resident microbiota play a significant role in combatting diseases and responding to stress. Hence, there is growing traction to build a knowledge base about this ecosystem, particularly to characterize the bidirectional relationship between the host and microbiota. In this context, metabolomics has emerged as the major converging node of this entire ecosystem. Systematic comprehension of this resourceful omics component can elucidate the organism-specific response trajectory and the communication grid across the ecosystem embodying meta-organisms. Translating this knowledge into designing nutraceuticals and next-generation therapy are ongoing. Its major hindrance is a significant knowledge gap about the underlying mechanisms maintaining a delicate balance within this ecosystem. To bridge this knowledge gap, a holistic picture of the available information has been presented with a primary focus on the microbiota-metabolite relationship dynamics. The central theme of this article is the gut-brain axis and the participating microbial metabolites that impact cerebral functions.
Collapse
Affiliation(s)
- Nabarun Chakraborty
- Medical Readiness Systems Biology, CMPN, WRAIR, Silver Spring, MD, United States
| |
Collapse
|
8
|
Mohr AE, Senkus KE, McDermid JM, Berger PK, Perrin MT, Handu D. Human Milk Nutrient Composition Data is Critically Lacking in the United States and Canada: Results from a Systematic Scoping Review of 2017-2022. Adv Nutr 2023; 14:1617-1632. [PMID: 37758059 PMCID: PMC10721511 DOI: 10.1016/j.advnut.2023.09.007] [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: 04/18/2023] [Revised: 09/14/2023] [Accepted: 09/19/2023] [Indexed: 10/03/2023] Open
Abstract
Characterization of the nutrients in human milk is important to understand the dietary and developmental requirements of infants. The objective of this review was to summarize the state-of-the-science on the nutrient composition of human milk in the United States and Canada published from 2017 to 2022. Four databases were searched for randomized controlled studies and others given the scoping nature of this review. We limited type to mature milk collected 21 d postpartum and beyond from lactating individuals in the United States and Canada who gave birth at 37-wk gestation or later (full-term). Outcomes of interest included traditional macro- and micronutrients, including human milk oligosaccharides (HMOs), and milk volume. The publication date range was selected as January 1, 2017, to the day the literature search was performed. A total of 32 articles were included in the scoping review from primarily longitudinal cohort or cross-sectional designs. The most prevalent sample collection method was full-breast expression (n = 20) with most studies (n = 26) collecting samples from a single timepoint. Carbohydrates (HMOs [n = 12], glucose [n = 8], and lactose [n = 6]) and protein (n = 5) were the most frequently assessed nutrients in this body of work, with consensus among studies that glucose is present in limited concentrations compared to lactose (24-64 mg/dL compared with 6-7 g/dL) and that HMOs are influenced by temporality and secretor status. Included studies displayed an overall level of heterogeneity and sparsity paralleling previous reports and nutrient data in the USDA FoodData Central system. Much of the data extracted from retained articles generally provided analysis of a specific nutrient or group of nutrients. Moreover, many studies did not use the preferred analytical methods as outlined by the Human Milk Composition Initiative to increase measurement confidence. Up-to-date nutrient composition data of human milk is still greatly needed as it is paramount for the management of infant feeding, assessment of infant and maternal nutritional and health needs, and as a reference for infant formula development.
Collapse
Affiliation(s)
- Alex E Mohr
- College of Health Solutions, Arizona State University, Phoenix, AZ, United States
| | - Katelyn E Senkus
- Department of Human Nutrition, The University of Alabama, Tuscaloosa, AL, United States
| | | | - Paige K Berger
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Boston, MA, United States
| | - Maryanne T Perrin
- Department of Nutrition, University of North Carolina Greensboro, Greensboro, NC, United States
| | - Deepa Handu
- Evidence Analysis Center, Academy of Nutrition and Dietetics, Chicago, IL, United States.
| |
Collapse
|
9
|
Abstract
Cardiometabolic disease comprises cardiovascular and metabolic dysfunction and underlies the leading causes of morbidity and mortality, both within the United States and worldwide. Commensal microbiota are implicated in the development of cardiometabolic disease. Evidence suggests that the microbiome is relatively variable during infancy and early childhood, becoming more fixed in later childhood and adulthood. Effects of microbiota, both during early development, and in later life, may induce changes in host metabolism that modulate risk mechanisms and predispose toward the development of cardiometabolic disease. In this review, we summarize the factors that influence gut microbiome composition and function during early life and explore how changes in microbiota and microbial metabolism influence host metabolism and cardiometabolic risk throughout life. We highlight limitations in current methodology and approaches and outline state-of-the-art advances, which are improving research and building toward refined diagnosis and treatment options in microbiome-targeted therapies.
Collapse
Affiliation(s)
- Curtis L Gabriel
- Division of Gastroenterology, Hepatology and Nutrition (C.L.G.), Vanderbilt University Medical Center, Nashville
- Tennessee Center for AIDS Research (C.L.G.), Vanderbilt University Medical Center, Nashville
| | - Jane F Ferguson
- Division of Cardiovascular Medicine (J.F.F.), Vanderbilt University Medical Center, Nashville
- Vanderbilt Microbiome Innovation Center (J.F.F.), Vanderbilt University Medical Center, Nashville
- Vanderbilt Institute for Infection, Immunology, and Inflammation (J.F.F.), Vanderbilt University Medical Center, Nashville
| |
Collapse
|
10
|
Baumgartel K, Stevens M, Vijayakumar N, Saint Fleur A, Prescott S, Groer M. The Human Milk Metabolome: A Scoping Literature Review. J Hum Lact 2023; 39:255-277. [PMID: 36924445 DOI: 10.1177/08903344231156449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
BACKGROUND Human milk is a complex source of nutrition and other bioactives that protects infants from disease, holding a lifetime of beneficial effects. The field of metabolomics provides a robust platform through which we can better understand human milk at a level rarely examined. RESEARCH AIM To Identify, describe, synthesize, and critically analyze the literature within the past 5 years related to the human milk metabolome. METHODS We conducted a scoping literature review and quality analysis of the recent science reflecting untargeted metabolomic approaches to examining human milk. We searched six databases using the terms "breast milk," "metabolome," "metabolite," and "human milk," Out of more than 1,069 abstracts, we screened and identified 22 articles that met our inclusion criteria. RESULTS We extracted data related to the study author, geographic location, research design, analyses, platform used, and results. We also extracted data related to human milk research activities, including collection protocol, infant/maternal considerations, and time. Selected studies focused on a variety of phenotypes, including maternal and infant disease. Investigators used varying approaches to evaluate the metabolome, and differing milk collection protocols were observed. CONCLUSION The human milk metabolome is informed by many factors-which may contribute to infant health outcomes-that have resulted in disparate milk metabolomic profiles. Standardized milk collection and storage procedures should be implemented to minimize degradation. Investigators may use our findings to develop research questions that test a targeted metabolomic approach.
Collapse
Affiliation(s)
| | - Monica Stevens
- College of Medicine, University of South Florida, Tampa, FL, USA
| | - Nisha Vijayakumar
- School of Public Health, University of South Florida, Tampa, FL, USA
| | | | | | - Maureen Groer
- College of Nursing, University of Tennessee, Knoxville, TN, USA
| |
Collapse
|
11
|
Couvillion SP, Mostoller KE, Williams JE, Pace RM, Stohel IL, Peterson HK, Nicora CD, Nakayasu ES, Webb-Robertson BJM, McGuire MA, McGuire MK, Metz TO. Interrogating the role of the milk microbiome in mastitis in the multi-omics era. Front Microbiol 2023; 14:1105675. [PMID: 36819069 PMCID: PMC9932517 DOI: 10.3389/fmicb.2023.1105675] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
There is growing interest in a functional understanding of milk-associated microbiota as there is ample evidence that host-associated microbial communities play an active role in host health and phenotype. Mastitis, characterized by painful inflammation of the mammary gland, is prevalent among lactating humans and agricultural animals and is associated with significant clinical and economic consequences. The etiology of mastitis is complex and polymicrobial and correlative studies have indicated alterations in milk microbial community composition. Recent evidence is beginning to suggest that a causal relationship may exist between the milk microbiota and host phenotype in mastitis. Multi-omic approaches can be leveraged to gain a mechanistic, molecular level understanding of how the milk microbiome might modulate host physiology, thereby informing strategies to prevent and ameliorate mastitis. In this paper, we review existing studies that have utilized omics approaches to investigate the role of the milk microbiome in mastitis. We also summarize the strengths and challenges associated with the different omics techniques including metagenomics, metatranscriptomics, metaproteomics, metabolomics and lipidomics and provide perspective on the integration of multiple omics technologies for a better functional understanding of the milk microbiome.
Collapse
Affiliation(s)
- Sneha P. Couvillion
- Pacific Northwest National Laboratory, Earth and Biological Sciences Directorate, Richland, WA, United States,*Correspondence: Sneha P. Couvillion, ✉
| | - Katie E. Mostoller
- Pacific Northwest National Laboratory, Earth and Biological Sciences Directorate, Richland, WA, United States
| | - Janet E. Williams
- Department of Animal, Veterinary, and Food Sciences, University of Idaho, Moscow, ID, United States
| | - Ryan M. Pace
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID, United States
| | - Izabel L. Stohel
- Pacific Northwest National Laboratory, Earth and Biological Sciences Directorate, Richland, WA, United States
| | - Haley K. Peterson
- Department of Animal, Veterinary, and Food Sciences, University of Idaho, Moscow, ID, United States
| | - Carrie D. Nicora
- Pacific Northwest National Laboratory, Earth and Biological Sciences Directorate, Richland, WA, United States
| | - Ernesto S. Nakayasu
- Pacific Northwest National Laboratory, Earth and Biological Sciences Directorate, Richland, WA, United States
| | - Bobbie-Jo M. Webb-Robertson
- Pacific Northwest National Laboratory, Earth and Biological Sciences Directorate, Richland, WA, United States
| | - Mark A. McGuire
- Department of Animal, Veterinary, and Food Sciences, University of Idaho, Moscow, ID, United States
| | - Michelle K. McGuire
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID, United States
| | - Thomas O. Metz
- Pacific Northwest National Laboratory, Earth and Biological Sciences Directorate, Richland, WA, United States,Thomas O. Metz, ✉
| |
Collapse
|
12
|
Lahdenperä M, Galante L, Gonzales-Inca C, Vahtera J, Pentti J, Rautava S, Käyhkö N, Yonemitsu C, Gupta J, Bode L, Lagström H. Residential green environments are associated with human milk oligosaccharide diversity and composition. Sci Rep 2023; 13:216. [PMID: 36604578 PMCID: PMC9816313 DOI: 10.1038/s41598-022-27317-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023] Open
Abstract
Increased exposure to greener environments has been suggested to lead to health benefits in children, but the associated mechanisms in early life, particularly via biological mediators such as altered maternal milk composition, remain largely unexplored. We investigated the associations between properties of the mother's residential green environment, measured as (1) greenness (Normalized Difference Vegetation index, NDVI), (2) Vegetation Cover Diversity (VCDI) and (3) Naturalness Index (NI), and human milk oligosaccharides (HMOs), known for their immune- and microbiota-related health effects on the infant (N = 795 mothers). We show that HMO diversity increases and concentrations of several individual HMOs and HMO groups change with increased VCDI and NI in residential green environments. This suggests that variation in residential green environments may influence the infant via maternal milk through modified HMO composition. The results emphasize the mediating role of breastfeeding between the residential green environments and health in early life.
Collapse
Affiliation(s)
- Mirkka Lahdenperä
- Department of Biology, University of Turku, Turku, Finland. .,Department of Public Health, University of Turku and Turku University Hospital, Turku, Finland. .,Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland.
| | - Laura Galante
- grid.1374.10000 0001 2097 1371Department of Biology, University of Turku, Turku, Finland ,grid.1374.10000 0001 2097 1371Department of Public Health, University of Turku and Turku University Hospital, Turku, Finland ,grid.1374.10000 0001 2097 1371Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland ,grid.4827.90000 0001 0658 8800School of Health and Social Care, Faculty of Medicine, Health and Life Sciences, Swansea University, Swansea, SA2 8PP UK
| | - Carlos Gonzales-Inca
- grid.1374.10000 0001 2097 1371Department of Geography and Geology, University of Turku, Turku, Finland
| | - Jussi Vahtera
- grid.1374.10000 0001 2097 1371Department of Public Health, University of Turku and Turku University Hospital, Turku, Finland ,grid.1374.10000 0001 2097 1371Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Jaana Pentti
- grid.1374.10000 0001 2097 1371Department of Public Health, University of Turku and Turku University Hospital, Turku, Finland ,grid.1374.10000 0001 2097 1371Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland ,grid.7737.40000 0004 0410 2071Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Samuli Rautava
- grid.1374.10000 0001 2097 1371Department of Pediatrics, University of Turku, Turku, Finland ,grid.7737.40000 0004 0410 2071Children’s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Niina Käyhkö
- grid.1374.10000 0001 2097 1371Department of Geography and Geology, University of Turku, Turku, Finland
| | - Chloe Yonemitsu
- grid.266100.30000 0001 2107 4242Department of Pediatrics and Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (MOMI CORE), University of California San Diego, La Jolla, CA USA
| | - Julia Gupta
- grid.266100.30000 0001 2107 4242Department of Pediatrics and Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (MOMI CORE), University of California San Diego, La Jolla, CA USA
| | - Lars Bode
- grid.266100.30000 0001 2107 4242Department of Pediatrics and Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (MOMI CORE), University of California San Diego, La Jolla, CA USA
| | - Hanna Lagström
- grid.1374.10000 0001 2097 1371Department of Public Health, University of Turku and Turku University Hospital, Turku, Finland ,grid.1374.10000 0001 2097 1371Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| |
Collapse
|
13
|
Lundgren SN, Madan JC, Karagas MR, Morrison HG, Christensen BC, Hoen AG. Human milk-associated bacterial communities associate with the infant gut microbiome over the first year of life. Front Microbiol 2023; 14:1164553. [PMID: 37138613 PMCID: PMC10149717 DOI: 10.3389/fmicb.2023.1164553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 03/27/2023] [Indexed: 05/05/2023] Open
Abstract
Introduction Microbial communities inhabiting the human infant gut are important for immune system development and lifelong health. One critical exposure affecting the bacterial colonization of the infant gut is consumption of human milk, which contains diverse microbial communities and prebiotics. We hypothesized that human milk-associated microbial profiles are associated with those of the infant gut. Methods Maternal-infant dyads enrolled in the New Hampshire Birth Cohort Study (n = 189 dyads) contributed breast milk and infant stool samples collected approximately at 6 weeks, 4 months, 6 months, 9 months, and 12 months postpartum (n = 572 samples). Microbial DNA was extracted from milk and stool and the V4-V5 region of the bacterial 16S rRNA gene was sequenced. Results Clustering analysis identified three breast milk microbiome types (BMTs), characterized by differences in Streptococcus, Staphylococcus, Pseudomonas, Acinetobacter, and microbial diversity. Four 6-week infant gut microbiome types (6wIGMTs) were identified, differing in abundances of Bifidobacterium, Bacteroides, Clostridium, Streptococcus, and Escherichia/Shigella, while two 12-month IGMTs (12mIGMTs) differed primarily by Bacteroides presence. At 6 weeks, BMT was associated with 6wIGMT (Fisher's exact test value of p = 0.039); this association was strongest among infants delivered by Cesarean section (Fisher's exact test value of p = 0.0028). The strongest correlations between overall breast milk and infant stool microbial community structures were observed when comparing breast milk samples to infant stool samples collected at a subsequent time point, e.g., the 6-week breast milk microbiome associated with the 6-month infant gut microbiome (Mantel test Z-statistic = 0.53, value of p = 0.001). Streptoccous and Veillonella species abundance were correlated in 6-week milk and infant stool, and 4- and 6-month milk Pantoea species were associated with infant stool Lachnospiraceae genera at 9 and 12 months. Discussion We identified clusters of human milk and infant stool microbial communities that were associated in maternal-infant dyads at 6 weeks of life and found that milk microbial communities were more strongly associated with infant gut microbial communities in infants delivered operatively and after a lag period. These results suggest that milk microbial communities have a long-term effect on the infant gut microbiome both through sharing of microbes and other molecular mechanisms.
Collapse
Affiliation(s)
- Sara N. Lundgren
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Juliette C. Madan
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
- Department of Pediatrics, Children’s Hospital at Dartmouth, Lebanon, NH, United States
- Department of Psychiatry, Children’s Hospital at Dartmouth, Lebanon, NH, United States
| | - Margaret R. Karagas
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
- Department of Community and Family Medicine, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Hilary G. Morrison
- Josephine Bay Paul Center, Marine Biological Laboratory, Woods Hole, MA, United States
| | - Brock C. Christensen
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
- Department of Community and Family Medicine, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
- *Correspondence: Brock C. Christensen,
| | - Anne G. Hoen
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| |
Collapse
|
14
|
Ahuja JKC, Casavale KO, Li Y, Hopperton KE, Chakrabarti S, Hines EP, Brooks SPJ, Bondy GS, MacFarlane AJ, Weiler HA, Wu X, Borghese MM, Ahluwalia N, Cheung W, Vargas AJ, Arteaga S, Lombo T, Fisher MM, Hayward D, Pehrsson PR. Perspective: Human Milk Composition and Related Data for National Health and Nutrition Monitoring and Related Research. Adv Nutr 2022; 13:2098-2114. [PMID: 36084013 PMCID: PMC9776678 DOI: 10.1093/advances/nmac099] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 04/18/2022] [Accepted: 09/07/2022] [Indexed: 01/28/2023] Open
Abstract
National health and nutrition monitoring is an important federal effort in the United States and Canada, and the basis for many of their nutrition and health policies. Understanding of child exposures through human milk (HM) remains out of reach due to lack of current and representative data on HM's composition and intake volume. This article provides an overview of the current national health and nutrition monitoring activities for HM-fed children, HM composition (HMC) and volume data used for exposure assessment, categories of potential measures in HM, and associated variability factors. In this Perspective, we advocate for a framework for collection and reporting of HMC data for national health and nutrition monitoring and programmatic needs, including a shared vision for a publicly available Human Milk Composition Data Repository (HMCD-R) to include essential metadata associated with HMC. HMCD-R can provide a central, integrated platform for researchers and public health officials for compiling, evaluating, and sharing HMC data. The compiled compositional and metadata in HMCD-R would provide pertinent measures of central tendency and variability and allow use of modeling techniques to approximate compositional profiles for subgroups, providing more accurate exposure assessments for purposes of monitoring and surveillance. HMC and related metadata could facilitate understanding the complexity and variability of HM composition, provide crucial data for assessment of infant and maternal nutritional needs, and inform public health policies, food and nutrition programs, and clinical practice guidelines.
Collapse
Affiliation(s)
- Jaspreet K C Ahuja
- Methods and Application of Food Composition Laboratory, Beltsville Human
Nutrition Research Center, Agricultural Research Services, US Department
of Agriculture, Beltsville, Maryland, USA
| | - Kellie O Casavale
- Center for Food Safety and Applied Nutrition, Food and Drug
Administration, US Department of Health and Human Services, College
Park, Maryland, USA
| | - Ying Li
- Methods and Application of Food Composition Laboratory, Beltsville Human
Nutrition Research Center, Agricultural Research Services, US Department
of Agriculture, Beltsville, Maryland, USA
| | - Kathryn E Hopperton
- Nutrition Premarket Assessment Division, Bureau of Nutritional Sciences,
Food Directorate, Health Products and Food Branch, Health Canada,
Ottawa, Ontario, Canada
| | - Subhadeep Chakrabarti
- Nutrition Premarket Assessment Division, Bureau of Nutritional Sciences,
Food Directorate, Health Products and Food Branch, Health Canada,
Ottawa, Ontario, Canada
| | - Erin P Hines
- Reproductive and Developmental Toxicology Branch, Public Health and
Integrated Toxicology Division, US Environmental Protection Agency,
Chapel Hill, North Carolina, USA
| | - Stephen P J Brooks
- Nutrition Research Division, Bureau of Nutritional Sciences, Food
Directorate, Health Products and Food Branch, Health Canada, Ottawa,
Ontario, Canada
| | - Genevieve S Bondy
- Bureau of Chemical Safety, Food Directorate, Health Products and Food
Branch, Health Canada, Ottawa, Ontario, Canada
| | - Amanda J MacFarlane
- Nutrition Research Division, Bureau of Nutritional Sciences, Food
Directorate, Health Products and Food Branch, Health Canada, Ottawa,
Ontario, Canada
| | - Hope A Weiler
- Nutrition Research Division, Bureau of Nutritional Sciences, Food
Directorate, Health Products and Food Branch, Health Canada, Ottawa,
Ontario, Canada
| | - Xianli Wu
- Methods and Application of Food Composition Laboratory, Beltsville Human
Nutrition Research Center, Agricultural Research Services, US Department
of Agriculture, Beltsville, Maryland, USA
| | - Michael M Borghese
- Environmental Health Sciences and Research Bureau, Health
Canada, Ottawa, Ontario, Canada
| | - Namanjeet Ahluwalia
- National Center for Health Statistics, Centers for Disease Control and
Prevention, Department of Health and Human Services, Hyattsville,
Maryland, USA
| | - Winnie Cheung
- Nutrition Premarket Assessment Division, Bureau of Nutritional Sciences,
Food Directorate, Health Products and Food Branch, Health Canada,
Ottawa, Ontario, Canada
| | - Ashley J Vargas
- Eunice Kennedy Shriver National Institute of Child Health and Human
Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Sonia Arteaga
- Environmental influences on Child Health Outcomes (ECHO) Program, Office of
the Director, National Institutes of Health, Bethesda, Maryland,
USA
| | - Tania Lombo
- Maternal Adolescent Pediatric Research Branch, Prevention Science Program,
Division of AIDS, National Institute of Allergy and Infectious Diseases
(NIAID), Bethesda, Maryland, USA
| | - Mandy M Fisher
- Environmental Health Sciences and Research Bureau, Health
Canada, Ottawa, Ontario, Canada
| | - Deborah Hayward
- Nutrition Premarket Assessment Division, Bureau of Nutritional Sciences,
Food Directorate, Health Products and Food Branch, Health Canada,
Ottawa, Ontario, Canada
| | - Pamela R Pehrsson
- Methods and Application of Food Composition Laboratory, Beltsville Human
Nutrition Research Center, Agricultural Research Services, US Department
of Agriculture, Beltsville, Maryland, USA
| |
Collapse
|
15
|
Kortesniemi M, Jafari T, Zhang Y, Yang B. 1H NMR Metabolomics of Chinese Human Milk at Different Stages of Lactation among Secretors and Non-Secretors. Molecules 2022; 27:molecules27175526. [PMID: 36080292 PMCID: PMC9458218 DOI: 10.3390/molecules27175526] [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: 07/17/2022] [Revised: 08/17/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Human milk is an intricate, bioactive food promoting infant health. We studied the composition of human milk samples collected over an 8-month lactation using 1H NMR metabolomics. A total of 72 human breast milk samples were collected from ten Chinese mothers at eight different time points. The concentrations of ten human milk oligosaccharides (HMOs), fucose and lactose were quantified. Six of the mothers were classified as Lewis-positive secretors (Se+Le+) and four as Lewis-positive non-secretors (Se−Le+) based on the levels of 2′-fucosyllactose (2′-FL), lacto-N-fucopentaose (LNFP) II, lactodifucotetraose (LDFT) and lacto-N-neotetraose (LNnT). Acetate, citrate, short/medium-chain fatty acids, glutamine and urea showed a time-dependent trend in relation to the stage of lactation. The concentrations of 2′-FL, 3-FL (3-fucosyllactose), 3′-SL (3′-sialyllactose), LDFT, LNFP I, LNFP II, LNFP III, LNnT, LNT (lacto-N-tetraose), and fucose were statistically different between secretors and non-secretors. A temporal difference of approximately 1–2 months between the development of non-secretor and secretor HMO profiles was shown. The results highlighted the importance of long-term breastfeeding, especially among non-secretors.
Collapse
Affiliation(s)
- Maaria Kortesniemi
- Food Sciences, Department of Life Technologies, University of Turku, FI-20014 Turku, Finland
- Correspondence: (M.K.); (Y.Z.)
| | - Tahereh Jafari
- Food Sciences, Department of Life Technologies, University of Turku, FI-20014 Turku, Finland
| | - Yumei Zhang
- Department of Nutrition and Food Hygiene, Peking University, Xueyuan Road 38, Haidian District, Beijing 100191, China
- Correspondence: (M.K.); (Y.Z.)
| | - Baoru Yang
- Food Sciences, Department of Life Technologies, University of Turku, FI-20014 Turku, Finland
| |
Collapse
|
16
|
Poulsen KO, Meng F, Lanfranchi E, Young JF, Stanton C, Ryan CA, Kelly AL, Sundekilde UK. Dynamic Changes in the Human Milk Metabolome Over 25 Weeks of Lactation. Front Nutr 2022; 9:917659. [PMID: 35911093 PMCID: PMC9331903 DOI: 10.3389/fnut.2022.917659] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/13/2022] [Indexed: 12/11/2022] Open
Abstract
Human milk (HM) provides essential nutrition for ensuring optimal infant growth and development postpartum. Metabolomics offers insight into the dynamic composition of HM. Studies have reported the impact of lactation stage, maternal genotype, and gestational age on HM metabolome. However, the majority of the studies have considered changes within the first month of lactation or sampled with large intervals. This leaves a gap in the knowledge of progressing variation in HM composition beyond the first month of lactation. The objective of this study was to investigate whether the HM metabolome from mothers with term deliveries varies beyond 1 month of lactation, during the period in which HM is considered fully mature. Human milk samples (n = 101) from 59 mothers were collected at weeks 1-2, 3-5, 7-9, and 20-25 postpartum and analyzed using 1H nuclear magnetic resonance spectroscopy. Several metabolites varied over lactation and exhibited dynamic changes between multiple time points. Higher levels of HM oligosaccharides, cis-aconitate, O-phosphocholine, O-acetylcarnitine, gluconate, and citric acid were observed in early lactation, whereas later in lactation, levels of lactose, 3-fucosyllactose, glutamine, glutamate, and short- and medium-chain fatty acids were increased. Notably, we demonstrate that the HM metabolome is dynamic during the period of maturity.
Collapse
Affiliation(s)
- Katrine Overgaard Poulsen
- Department of Food Science, Aarhus University, Aarhus, Denmark
- Sino-Danish Center for Education and Research, Aarhus, Denmark
| | - Fanyu Meng
- School of Food and Nutritional Sciences, University College Cork, Cork, Ireland
| | - Elisa Lanfranchi
- School of Food and Nutritional Sciences, University College Cork, Cork, Ireland
- ACIB – Austrian Centre of Industrial Biotechnology, Graz, Austria
| | | | | | - C. Anthony Ryan
- Brookfield School of Medicine and Health, University College Cork, Cork, Ireland
| | - Alan L. Kelly
- School of Food and Nutritional Sciences, University College Cork, Cork, Ireland
| | | |
Collapse
|
17
|
Selma-Royo M, Calvo-Lerma J, Bäuerl C, Esteban-Torres M, Cabrera-Rubio R, Collado MC. Human milk microbiota: what did we learn in the last 20 years? MICROBIOME RESEARCH REPORTS 2022; 1:19. [PMID: 38046359 PMCID: PMC10688795 DOI: 10.20517/mrr.2022.05] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/06/2022] [Accepted: 04/29/2022] [Indexed: 12/05/2023]
Abstract
Human milk (HM) is the gold standard for infant nutrition during the first months of life. Beyond its nutritional components, its complex bioactive composition includes microorganisms, their metabolites, and oligosaccharides, which also contribute to gut colonization and immune system maturation. There is growing evidence of the beneficial effects of bacteria present in HM. However, current research presents limited data on the presence and functions of other organisms. The potential biological impacts on maternal and infant health outcomes, the factors contributing to milk microbes' variations, and the potential functions in the infant's gut remain unclear. This review provides a global overview of milk microbiota, what the actual knowledge is, and what the gaps and challenges are for the next years.
Collapse
Affiliation(s)
| | | | | | | | | | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia 46980, Spain
| |
Collapse
|
18
|
Saladrigas-García M, Solà-Oriol D, López-Vergé S, D'Angelo M, Carmen Collado M, Nielsen B, Faldyna M, Francisco Pérez J, Martín-Orúe SM. Potential effect of two Bacillus probiotic strains on performance and fecal microbiota of breeding sows and their piglets. J Anim Sci 2022; 100:6580401. [PMID: 35512239 PMCID: PMC9175292 DOI: 10.1093/jas/skac163] [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: 01/13/2022] [Accepted: 04/29/2022] [Indexed: 11/19/2022] Open
Abstract
The effect of long-term administration of two Bacillus strains was tested on 98 breeding sows and their litters allotted into three treatments: a control group (CON); supplemented with 5 × 108 cfu/kg B. subtilis − 541 (BSU); or with 5 × 108 cfu/kg B. amyloliquefaciens − 516 (BAM). Reproductive and performance variables were recorded over three cycles with 56 dams remaining through the third lactation. Blood and fecal samples were taken longitudinally from 12 sows per treatment on days 8 and 21 of the third lactation and milk samples were taken on day 21. Feces from one piglet per litter was sampled on days 21 and 33 and jejunal gene expression was assessed in two piglets on day 21. Changes in fecal microbiota were assessed by 16S rRNA gene sequencing (Illumina MiSeq) and gene expression by Open-Array technology. Metabolomic responses were analyzed in milk by NMR and Ig-G and Ig-A specific antibodies were determined by ELISA. No significant differences were observed on feed intake, body weight, or fat mobilization of the sows. However, a significant increase in the total number of piglets born was observed in supplemented sows. Although the increase was seen from the first cycle with BAM, improvements were not seen with BSU until the third cycle. BAM also increased the number of born-alive and weaned piglets. NMR analysis showed an impact of BAM on milk composition. No differences were found in milk or blood immunoglobulins. A different structure of the fecal microbiota was found in supplemented sows, with changes across phylum, family, and genus. These changes were greater at day 8, suggesting a relevant role of probiotics establishing a new intestinal balance after labor. Shifts in the microbiota were also seen in the piglets, with a clearer impact post-weaning than in suckling. In this regard, correlations between microbial groups of sows and piglets showed a higher link with weaned (d33) than with suckling pigs (d21), reinforcing the idea of an early maternal carry-over. No changes due to treatment in jejunal gene expression were detected; however, piglet size had a clear impact on different genes. In summary, the addition of both probiotics, and particularly Bacillus amyloliquefaciens, demonstrated potential benefits on the prolificacy of sows. Daily feeding of Bacillus amyloliquefaciens resulted in an increase in the number of weaned piglets. The high correlations between the compositions of the microbiota of sows and their piglets are evidence of maternal imprinting, with effects lasting beyond weaning.
Collapse
Affiliation(s)
- Mireia Saladrigas-García
- Animal Nutrition and Welfare Service, Department of Animal and Food Sciences, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - David Solà-Oriol
- Animal Nutrition and Welfare Service, Department of Animal and Food Sciences, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Sergi López-Vergé
- Animal Nutrition and Welfare Service, Department of Animal and Food Sciences, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Matilde D'Angelo
- Animal Nutrition and Welfare Service, Department of Animal and Food Sciences, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Valencia 49860, Spain
| | | | - Martin Faldyna
- Veterinary Research Institute, Brno 62132, Czech Republic
| | - José Francisco Pérez
- Animal Nutrition and Welfare Service, Department of Animal and Food Sciences, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Susana M Martín-Orúe
- Animal Nutrition and Welfare Service, Department of Animal and Food Sciences, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| |
Collapse
|
19
|
Niewiem M, Grzybowska-Chlebowczyk U. Intestinal Barrier Permeability in Allergic Diseases. Nutrients 2022; 14:nu14091893. [PMID: 35565858 PMCID: PMC9101724 DOI: 10.3390/nu14091893] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/25/2022] [Accepted: 04/28/2022] [Indexed: 02/01/2023] Open
Abstract
The role of intestinal permeability (IP) markers among children and adults with food allergies is not fully understood, and the identification of biological indicators/markers that predict growth retardation in children with allergic diseases and atopy has not been well explained. Studies have shown that patients with atopic diseases respond abnormally to food allergens. Accordingly, differences in the types of immune complexes formed in response to antigen challenges are significant, which seems to underlie the systemic signs of the food allergy. Increased intestinal permeability over the course of a food allergy allows allergens to penetrate through the intestinal barrier and stimulate the submucosal immune system. Additionally, the release of cytokines and inflammatory mediators enhances the degradation of the epithelial barrier and leads to an improper cycle, resulting in increased intestinal permeability. Several studies have also demonstrated increased permeability of the epithelial cells in those afflicted with atopic eczema and bronchial asthma. Ongoing research is aimed at finding various indicators to assess IP in patients with atopic diseases.
Collapse
|
20
|
Unexpected content of kynurenine in mother's milk and infant formulas. Sci Rep 2022; 12:6464. [PMID: 35440600 PMCID: PMC9018775 DOI: 10.1038/s41598-022-10075-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 03/14/2022] [Indexed: 11/08/2022] Open
Abstract
Mother's milk is widely recommended as complete food for the offspring in earliest postnatal time. However, the knowledge about detailed composition and the physiological role of bioactive components of breast milk is incomplete. Therefore, the aim of our study was to determine the content of kynurenine (KYN) in human breast milk during lactation and to explore the effects exerted by intragastric KYN administration from birth to weaning on physical and psychomotor development of adult rats. We found that KYN is consistently present in human milk and its content gradually increased from day 4 to 28 after delivery and that it is present in commercial baby formulas in amounts noticeably exceeding its physiological range. Animal studies showed that KYN supplementation resulted in a marked elevation of absorptive surface of rat intestine and in enhanced expression of both, aryl hydrocarbon receptor and G protein-coupled receptor 35 in the intestinal tissue in rats. Moreover, we discovered that KYN administration from birth to weaning resulted in neurobehavioral changes in adult rats. Therefore, we postulate that further research is required to thoroughly understand the function of KYN in early developmental stages of mammals and to ensure the safety of its presence in baby food products.
Collapse
|
21
|
de Weerth C, Aatsinki AK, Azad MB, Bartol FF, Bode L, Collado MC, Dettmer AM, Field CJ, Guilfoyle M, Hinde K, Korosi A, Lustermans H, Mohd Shukri NH, Moore SE, Pundir S, Rodriguez JM, Slupsky CM, Turner S, van Goudoever JB, Ziomkiewicz A, Beijers R. Human milk: From complex tailored nutrition to bioactive impact on child cognition and behavior. Crit Rev Food Sci Nutr 2022; 63:7945-7982. [PMID: 35352583 DOI: 10.1080/10408398.2022.2053058] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human milk is a highly complex liquid food tailor-made to match an infant's needs. Beyond documented positive effects of breastfeeding on infant and maternal health, there is increasing evidence that milk constituents also impact child neurodevelopment. Non-nutrient milk bioactives would contribute to the (long-term) development of child cognition and behavior, a process termed 'Lactocrine Programming'. In this review we discuss the current state of the field on human milk composition and its links with child cognitive and behavioral development. To promote state-of-the-art methodologies and designs that facilitate data pooling and meta-analytic endeavors, we present detailed recommendations and best practices for future studies. Finally, we determine important scientific gaps that need to be filled to advance the field, and discuss innovative directions for future research. Unveiling the mechanisms underlying the links between human milk and child cognition and behavior will deepen our understanding of the broad functions of this complex liquid food, as well as provide necessary information for designing future interventions.
Collapse
Affiliation(s)
- Carolina de Weerth
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, EN Nijmegen, The Netherlands
| | - Anna-Katariina Aatsinki
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
| | - Meghan B Azad
- Department of Pediatrics and Child Health, Manitoba Interdisciplinary Lactation Centre, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Frank F Bartol
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
| | - Lars Bode
- Department of Pediatrics and Mother-Milk-Infant Center of Research Excellence (MOMI CORE), University of California San Diego, La Jolla, California, USA
| | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Paterna, Valencia, Spain
| | - Amanda M Dettmer
- Yale Child Study Center, Yale School of Medicine, New Haven, Connecticut, USA
| | - Catherine J Field
- Department of Agricultural, Food and Nutritional Science, College of Basic and Applied Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Meagan Guilfoyle
- Department of Anthropology, Indiana University, Bloomington, Indiana, USA
| | - Katie Hinde
- School of Human Evolution and Social Change, Arizona State University, Tempe, Arizona, USA
| | - Aniko Korosi
- Swammerdam Institute for Life Sciences, Center for Neuroscience, Brain Plasticity group, University of Amsterdam, Amsterdam, The Netherlands
| | - Hellen Lustermans
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, EN Nijmegen, The Netherlands
| | - Nurul Husna Mohd Shukri
- Department of Nutrition, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| | - Sophie E Moore
- Department of Women & Children's Health, King's College London, St Thomas' Hospital, London, UK
- School of Hygiene and Tropical Medicine, Nutrition Theme, MRC Unit The Gambia and the London, Fajara, The GambiaBanjul
| | - Shikha Pundir
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Juan Miguel Rodriguez
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| | - Carolyn M Slupsky
- Department of Nutrition and Department of Food Science and Technology, University of California, Davis, California, USA
| | - Sarah Turner
- Department of Community Health Sciences, Manitoba Interdisciplinary Lactation Centre, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Johannes B van Goudoever
- Department of Pediatrics, Amsterdam UMC, University of Amsterdam, Vrije Universiteit, Emma Children's Hospital, Amsterdam, The Netherlands
| | - Anna Ziomkiewicz
- Department of Anthropology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Roseriet Beijers
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, EN Nijmegen, The Netherlands
- Department of Social Development, Behavioural Science Institute, Radboud University, Nijmegen, The Netherlands
| |
Collapse
|
22
|
Human Milk Oligosaccharides and Bacterial Profile Modulate Infant Body Composition during Exclusive Breastfeeding. Int J Mol Sci 2022; 23:ijms23052865. [PMID: 35270006 PMCID: PMC8911220 DOI: 10.3390/ijms23052865] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/23/2022] [Accepted: 03/02/2022] [Indexed: 02/06/2023] Open
Abstract
Human milk is a complex and variable ecosystem fundamental to the development of newborns. This study aimed to investigate relationships between human milk oligosaccharides (HMO) and human milk bacterial profiles and infant body composition. Human milk samples (n = 60) were collected at two months postpartum. Infant and maternal body composition was measured with bioimpedance spectroscopy. Human milk bacterial profiles were assessed using full-length 16S rRNA gene sequencing and 19 HMOs were quantitated using high-performance liquid chromatography. Relative abundance of human milk bacterial taxa were significantly associated with concentrations of several fucosylated and sialylated HMOs. Individual human milk bacteria and HMO intakes and concentrations were also significantly associated with infant anthropometry, fat-free mass, and adiposity. Furthermore, when data were stratified based on maternal secretor status, some of these relationships differed significantly among infants born to secretor vs non-secretor mothers. In conclusion, in this pilot study the human milk bacterial profile and HMO intakes and concentrations were significantly associated with infant body composition, with associations modified by secretor status. Future research designed to increase the understanding of the mechanisms by which HMO and human milk bacteria modulate infant body composition should include intakes in addition to concentrations.
Collapse
|
23
|
Peila C, Sottemano S, Cesare Marincola F, Stocchero M, Pusceddu NG, Dessì A, Baraldi E, Fanos V, Bertino E. NMR Metabonomic Profile of Preterm Human Milk in the First Month of Lactation: From Extreme to Moderate Prematurity. Foods 2022; 11:foods11030345. [PMID: 35159496 PMCID: PMC8834565 DOI: 10.3390/foods11030345] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 02/06/2023] Open
Abstract
Understanding the composition of human milk (HM) can provide important insights into the links between infant nutrition, health, and development. In the present work, we have longitudinally investigated the metabolome of milk from 36 women delivering preterm at different gestational ages (GA): extremely (<28 weeks GA), very (29–31 weeks GA) or moderate (32–34 weeks GA) premature. Milk samples were collected at three lactation stages: colostrum (3–6 days post-partum), transitional milk (7–15 days post-partum) and mature milk (16–26 days post-partum). Multivariate and univariate statistical data analyses were performed on the 1H NMR metabolic profiles of specimens in relation to the degree of prematurity and lactation stage. We observed a high impact of both the mother’s phenotype and lactation time on HM metabolome composition. Furthermore, statistically significant differences, although weak, were observed in terms of GA when comparing extremely and moderately preterm milk. Overall, our study provides new insights into preterm HM metabolome composition that may help to optimize feeding of preterm newborns, and thus improve the postnatal growth and later health outcomes of these fragile patients.
Collapse
Affiliation(s)
- Chiara Peila
- Neonatal Unit, University of Turin, City of Health and Science of Turin, 10126 Turin, Italy; (C.P.); (S.S.); (E.B.)
| | - Stefano Sottemano
- Neonatal Unit, University of Turin, City of Health and Science of Turin, 10126 Turin, Italy; (C.P.); (S.S.); (E.B.)
| | - Flaminia Cesare Marincola
- Department of Chemical and Geological Sciences, Cittadella Universitaria di Monserrato, University of Cagliari, Monserrato, 09042 Cagliari, Italy;
- Correspondence: (F.C.M.); (M.S.)
| | - Matteo Stocchero
- Department of Women’s and Children’s Health, University of Padova, 35128 Padova, Italy;
- Institute of Pediatric Research (IRP), Fondazione Città della Speranza, 35128 Padova, Italy
- Correspondence: (F.C.M.); (M.S.)
| | - Nicoletta Grazia Pusceddu
- Department of Chemical and Geological Sciences, Cittadella Universitaria di Monserrato, University of Cagliari, Monserrato, 09042 Cagliari, Italy;
| | - Angelica Dessì
- Neonatal Intensive Care Unit, Neonatal Pathology and Neonatal Section, Azienda University Polyclinic, University of Cagliari, 09042 Cagliari, Italy; (A.D.); (V.F.)
| | - Eugenio Baraldi
- Department of Women’s and Children’s Health, University of Padova, 35128 Padova, Italy;
- Institute of Pediatric Research (IRP), Fondazione Città della Speranza, 35128 Padova, Italy
| | - Vassilios Fanos
- Neonatal Intensive Care Unit, Neonatal Pathology and Neonatal Section, Azienda University Polyclinic, University of Cagliari, 09042 Cagliari, Italy; (A.D.); (V.F.)
| | - Enrico Bertino
- Neonatal Unit, University of Turin, City of Health and Science of Turin, 10126 Turin, Italy; (C.P.); (S.S.); (E.B.)
| |
Collapse
|
24
|
A Comparison of Mother’s Milk and the Neonatal Urine Metabolome: A Unique Fingerprinting for Different Nutritional Phenotypes. Metabolites 2022; 12:metabo12020113. [PMID: 35208187 PMCID: PMC8879468 DOI: 10.3390/metabo12020113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/18/2022] [Accepted: 01/22/2022] [Indexed: 12/14/2022] Open
Abstract
The ability of metabolomics to provide a snapshot of an individual’s metabolic state makes it a very useful technique in neonatology for investigating the complex relationship between nutrition and the state of health of the newborn. Through an 1H-NMR metabolomics analysis, we aimed to investigate the metabolic profile of newborns by analyzing both urine and milk samples in relation to the birth weight of neonates classified as AGA (adequate for the gestational age, n = 51), IUGR (intrauterine growth restriction, n = 14), and LGA (large for gestational age, n = 15). Samples were collected at 7 ± 2 days after delivery. Of these infants, 42 were exclusively breastfed, while 38 received mixed feeding with a variable amount of commercial infant formula (less than 40%) in addition to breast milk. We observed a urinary spectral pattern for oligosaccharides very close to that of the corresponding mother’s milk in the case of exclusively breastfed infants, thus mirroring the maternal phenotype. The absence of this good match between the infant urine and human milk spectra in the case of mixed-fed infants could be reasonably ascribed to the use of a variable amount of commercial infant formulas (under 40%) added to breast milk. Furthermore, our findings did not evidence any significant differences in the spectral profiles in terms of the neonatal customize centile, i.e., AGA (adequate for gestational age), LGA (large for gestational age), or IGUR (intrauterine growth restriction). It is reasonable to assume that maternal human milk oligosaccharide (HMO) production is not or is only minimally influenced by the fetal growth conditions for unknown reasons. This hypothesis may be supported by our metabolomics-based results, confirming once again the importance of this approach in the neonatal field.
Collapse
|
25
|
The hidden universe of human milk microbiome: origin, composition, determinants, role, and future perspectives. Eur J Pediatr 2022; 181:1811-1820. [PMID: 35124754 PMCID: PMC9056486 DOI: 10.1007/s00431-022-04383-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 12/14/2022]
Abstract
UNLABELLED Although traditionally considered sterile, human milk is currently recognized as an alive ecosystem that harbors not only bacteria, but also viruses, fungi and yeasts, and minor genera, collectively known as the human milk microbiome (HMM). The seeding of HMM is a complex phenomenon whose dynamics are still a matter of research. Many factors contribute to its determination, both maternal, neonatal, environmental, and related to human milk itself. The transmission of microorganisms to the infant through breastfeeding may impact its present and future health, mainly shaping the GI tract microbiome and immune system. The existence and persistence of HMM as a conserved feature among different species may also have an evolutionary meaning, which will become apparent only in evolutionary times. CONCLUSION The complexities of HMM warrant further research in order to deepen our knowledge on its origin, determinants, and impact on infants' health. The practical and translational implications of research on HMM (e.g., reconstitution of donor human milk through inoculation of infant's own mother milk, modulation of HMM through maternal dietary supplementation) should not be overlooked. WHAT IS KNOWN • Human milk harbors a wide variety of microorganisms, ranging from bacteria to viruses, fungi and yeasts, and minor genera. • Human milk microbiome is shaped over time by many factors: maternal, neonatal, environmental, and related to human milk itself. • The transmission of microorganisms through breastfeeding may impact the infant's present and future health. WHAT IS NEW • We provide an overview on human milk microbiome, hopefully encouraging physicians to consider it among the other better-known breastfeeding benefits. • Further studies, with standardized and rigorous study designs to enhance accuracy and reproducibility of the results, are needed to deepen our knowledge of the human milk microbiota and its role in newborn and infant's health.
Collapse
|
26
|
Allali I, Abotsi RE, Tow LA, Thabane L, Zar HJ, Mulder NM, Nicol MP. Human microbiota research in Africa: a systematic review reveals gaps and priorities for future research. MICROBIOME 2021; 9:241. [PMID: 34911583 PMCID: PMC8672519 DOI: 10.1186/s40168-021-01195-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 11/14/2021] [Indexed: 05/11/2023]
Abstract
BACKGROUND The role of the human microbiome in health and disease is an emerging and important area of research; however, there is a concern that African populations are under-represented in human microbiome studies. We, therefore, conducted a systematic survey of African human microbiome studies to provide an overview and identify research gaps. Our secondary objectives were: (i) to determine the number of peer-reviewed publications; (ii) to identify the extent to which the researches focused on diseases identified by the World Health Organization [WHO] State of Health in the African Region Report as being the leading causes of morbidity and mortality in 2018; (iii) to describe the extent and pattern of collaborations between researchers in Africa and the rest of the world; and (iv) to identify leadership and funders of the studies. METHODOLOGY We systematically searched Medline via PubMed, Scopus, CINAHL, Academic Search Premier, Africa-Wide Information through EBSCOhost, and Web of Science from inception through to 1st April 2020. We included studies that characterized samples from African populations using next-generation sequencing approaches. Two reviewers independently conducted the literature search, title and abstract, and full-text screening, as well as data extraction. RESULTS We included 168 studies out of 5515 records retrieved. Most studies were published in PLoS One (13%; 22/168), and samples were collected from 33 of the 54 African countries. The country where most studies were conducted was South Africa (27/168), followed by Kenya (23/168) and Uganda (18/168). 26.8% (45/168) focused on diseases of significant public health concern in Africa. Collaboration between scientists from the United States of America and Africa was most common (96/168). The first and/or last authors of 79.8% of studies were not affiliated with institutions in Africa. Major funders were the United States of America National Institutes of Health (45.2%; 76/168), Bill and Melinda Gates Foundation (17.8%; 30/168), and the European Union (11.9%; 20/168). CONCLUSIONS There are significant gaps in microbiome research in Africa, especially those focusing on diseases of public health importance. There is a need for local leadership, capacity building, intra-continental collaboration, and national government investment in microbiome research within Africa. Video Abstract.
Collapse
Affiliation(s)
- Imane Allali
- Computational Biology Division, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, and Genomic Centre of Human Pathologies, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Regina E Abotsi
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, Cape Town, South Africa
- Department of Pharmaceutical Microbiology, School of Pharmacy, University of Health and Allied Sciences, Ho, Ghana
| | - Lemese Ah Tow
- Division of Medical Microbiology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Lehana Thabane
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
- Biostatistics Unit, Father Sean O'Sullivan Research Centre, St Joseph's Healthcare, Hamilton, Ontario, Canada
- Departments of Paediatrics and Anaesthesia, McMaster University, Hamilton, Ontario, Canada
- Centre for Evaluation of Medicine, St Joseph's Healthcare, Hamilton, Ontario, Canada
- Population Health Research Institute, Hamilton Health Sciences, Hamilton, Ontario, Canada
- Centre for Evidence-based Health Care, Faculty of Health Sciences, Stellenbosch University, Tygerberg, South Africa
- Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Heather J Zar
- Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, Cape Town, South Africa
- MRC Unit on Child & Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Nicola M Mulder
- Computational Biology Division, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Mark P Nicol
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
- Division of Medical Microbiology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
- School of Biomedical Sciences, University of Western Australia, M504, Perth, WA, 6009, Australia.
| |
Collapse
|
27
|
Abstract
The neonatal body provides a range of potential habitats, such as the gut, for microbes. These sites eventually harbor microbial communities (microbiotas). A "complete" (adult) gut microbiota is not acquired by the neonate immediately after birth. Rather, the exclusive, milk-based nutrition of the infant encourages the assemblage of a gut microbiota of low diversity, usually dominated by bifidobacterial species. The maternal fecal microbiota is an important source of bacterial species that colonize the gut of infants, at least in the short-term. However, development of the microbiota is influenced by the use of human milk (breast feeding), infant formula, preterm delivery of infants, caesarean delivery, antibiotic administration, family details and other environmental factors. Following the introduction of weaning (complementary) foods, the gut microbiota develops in complexity due to the availability of a diversity of plant glycans in fruits and vegetables. These glycans provide growth substrates for the bacterial families (such as members of the Ruminococcaceae and Lachnospiraceae) that, in due course, will dominate the gut microbiota of the adult. Although current data are often fragmentary and observational, it can be concluded that the nutrition that a child receives in early life is likely to impinge not only on the development of the microbiota at that time but also on the subsequent lifelong, functional relationships between the microbiota and the human host. The purpose of this review, therefore, is to discuss the importance of promoting the assemblage of functionally robust gut microbiotas at appropriate times in early life.
Collapse
Affiliation(s)
- Gerald W. Tannock
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
28
|
Selma-Royo M, Calvo Lerma J, Cortés-Macías E, Collado MC. Human milk microbiome: From actual knowledge to future perspective. Semin Perinatol 2021; 45:151450. [PMID: 34274151 DOI: 10.1016/j.semperi.2021.151450] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human milk is the gold standard for infant nutrition during the first months of life since it is perfectly adapted to the neonatal nutritional requirements and supports infant growth and development. Human milk contains a complex nutritional and bioactive composition including microorganisms and oligosaccharides which would also contribute to the gut and immune system maturation. Despite the growing evidence, the factors contributing to milk microbes' variations and the potential functions on the infant's gut are still uncovered. This short-review provides a general overview of milk microbiota, potential factors shaping its composition, contribution to the infant microbiota and immune system development, including the suggested biological relevance for infant health as well as the description of tools and strategies aimed to restore and module microbes in milk.
Collapse
Affiliation(s)
- Marta Selma-Royo
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia (Spain).
| | - Joaquim Calvo Lerma
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia (Spain)
| | - Erika Cortés-Macías
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia (Spain)
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia (Spain).
| |
Collapse
|
29
|
Human Milk Oligosaccharides: A Comprehensive Review towards Metabolomics. CHILDREN-BASEL 2021; 8:children8090804. [PMID: 34572236 PMCID: PMC8465502 DOI: 10.3390/children8090804] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/06/2021] [Accepted: 09/10/2021] [Indexed: 12/11/2022]
Abstract
Human milk oligosaccharides (HMOs) are the third most represented component in breast milk. They serve not only as prebiotics but they exert a protective role against some significant neonatal pathologies such as necrotizing enterocolitis. Furthermore, they can program the immune system and consequently reduce allergies and autoimmune diseases’ incidence. HMOs also play a crucial role in brain development and in the gut barrier’s maturation. Moreover, the maternal genetic factors influencing different HMO patterns and their modulation by the interaction and the competition between active enzymes have been widely investigated in the literature, but there are few studies concerning the role of other factors such as maternal health, nutrition, and environmental influence. In this context, metabolomics, one of the newest “omics” sciences that provides a snapshot of the metabolites present in bio-fluids, such as breast milk, could be useful to investigate the HMO content in human milk. The authors performed a review, from 2012 to the beginning of 2021, concerning the application of metabolomics to investigate the HMOs, by using Pubmed, Researchgate and Scopus as source databases. Through this technology, it is possible to know in real-time whether a mother produces a specific oligosaccharide, keeping into consideration that there are other modifiable and unmodifiable factors that influence HMO production from a qualitative and a quantitative point of view. Although further studies are needed to provide clinical substantiation, in the future, thanks to metabolomics, this could be possible by using a dipstick and adding the eventual missing oligosaccharide to the breast milk or formula in order to give the best and the most personalized nutritional regimen for each newborn, adjusting to different necessities.
Collapse
|
30
|
Sari RN, Pan J, Zhang W, Li Y, Zhu H, Pang X, Zhang S, Jiang S, Lu J, Lv J. Comparative Proteomics of Human Milk From Eight Cities in China During Six Months of Lactation in the Chinese Human Milk Project Study. Front Nutr 2021; 8:682429. [PMID: 34458300 PMCID: PMC8387594 DOI: 10.3389/fnut.2021.682429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/14/2021] [Indexed: 12/23/2022] Open
Abstract
Human milk (HM) is the golden standard of infant nutrition that can protect immature body function and enhance nutrition metabolism to ensure infant growth. Region specificity and lactation period could change the protein composition in HM. In this research, proteomics analysis was used to compare proteomes across eight cities, namely Harbin, Lanzhou, Guangzhou, Chengdu, Jinhua, Weihai, Zhengzhou, and Beijing, which represented the northeast, northwest, southeast, southwest, east, and north and central regions of China,. Proteins varied significantly among the cities. These different proteins were mainly involved in the process of platelet degranulation, innate immune response, and triglyceride metabolic process, which might be due to different living environments. These differences also lead to variation in protection and fat metabolism from mothers to infants in different cities. Four proteins were expressed differently during 6 months of lactation, namely Dipeptidyl peptidase 1, Lysozyme C, Carbonic anhydrase 6, and Chordin-like protein 2. The changes in these proteins might be because of the change of growth needs of the infants. The findings from our results might help to improve the understanding of HM as well as to design infant formula.
Collapse
Affiliation(s)
- Ratna Nurmalita Sari
- Institute of Food Science and Technology, Chinese Academy of Agricultural Science, Beijing, China
| | - Jiancun Pan
- Nutrition and Metabolism Research Division, Innovation Center, Heilongjiang Feihe Dairy Co., Ltd., Beijing, China.,PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, Beijing, China
| | - Wenyuan Zhang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Science, Beijing, China
| | - Yuanyuan Li
- Nutrition and Metabolism Research Division, Innovation Center, Heilongjiang Feihe Dairy Co., Ltd., Beijing, China.,PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, Beijing, China
| | - Huiquan Zhu
- Institute of Food Science and Technology, Chinese Academy of Agricultural Science, Beijing, China
| | - Xiaoyang Pang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Science, Beijing, China
| | - Shuwen Zhang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Science, Beijing, China
| | - Shilong Jiang
- Nutrition and Metabolism Research Division, Innovation Center, Heilongjiang Feihe Dairy Co., Ltd., Beijing, China.,PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, Beijing, China
| | - Jing Lu
- Institute of Food Science and Technology, Chinese Academy of Agricultural Science, Beijing, China.,School of Food and Health, Beijing Business and Technology University, Beijing, China
| | - Jiaping Lv
- Institute of Food Science and Technology, Chinese Academy of Agricultural Science, Beijing, China
| |
Collapse
|
31
|
Thum C, Wall CR, Weiss GA, Wang W, Szeto IMY, Day L. Changes in HMO Concentrations throughout Lactation: Influencing Factors, Health Effects and Opportunities. Nutrients 2021; 13:2272. [PMID: 34209241 PMCID: PMC8308359 DOI: 10.3390/nu13072272] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/28/2021] [Accepted: 06/28/2021] [Indexed: 12/18/2022] Open
Abstract
Human milk oligosaccharides (HMOs) are important functional biomolecules in human breast milk. Understanding the factors influencing differences in HMO composition and changes in their concentration over lactation can help to design feeding strategies that are well-adapted to infant's needs. This review summarises the total and individual concentration of HMOs from data published from 1999 to 2019. Studies show that the HMO concentrations are highest in colostrum (average 9-22 g/L), followed by slightly lower concentrations in transitional milk (average 8-19 g/L), with a gradual decline in mature milk as lactation progresses, from 6-15 g/L in breast milk collected within one month of birth, to 4-6 g/L after 6 months. Significant differences in HMO composition have been described between countries. Different HMOs were shown to be predominant over the course of lactation, e.g., 3-fucosyllactose increased over lactation, whereas 2'-fucosyllactose decreased. Recent clinical studies on infant formula supplemented with 2'-fucosyllactose in combination with other oligosaccharides showed its limited beneficial effect on infant health.
Collapse
Affiliation(s)
- Caroline Thum
- AgResearch Ltd., Te Ohu Rangahau Kai, Palmerston North 4474, New Zealand;
| | - Clare Rosemary Wall
- Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1142, New Zealand;
| | | | - Wendan Wang
- Yili Maternal and Infant Nutrition Institute, Inner Mongolia Yili Industrial Group, Co., Ltd., Fengtai District, Beijing 100071, China; (W.W.); (I.M.-Y.S.)
| | - Ignatius Man-Yau Szeto
- Yili Maternal and Infant Nutrition Institute, Inner Mongolia Yili Industrial Group, Co., Ltd., Fengtai District, Beijing 100071, China; (W.W.); (I.M.-Y.S.)
| | - Li Day
- AgResearch Ltd., Te Ohu Rangahau Kai, Palmerston North 4474, New Zealand;
| |
Collapse
|
32
|
Newton JM, Betts EL, Yiangou L, Ortega Roldan J, Tsaousis AD, Thompson GS. Establishing a Metabolite Extraction Method to Study the Metabolome of Blastocystis Using NMR. Molecules 2021; 26:molecules26113285. [PMID: 34072445 PMCID: PMC8199492 DOI: 10.3390/molecules26113285] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/24/2021] [Accepted: 05/26/2021] [Indexed: 02/06/2023] Open
Abstract
Blastocystis is an opportunistic parasite commonly found in the intestines of humans and other animals. Despite its high prevalence, knowledge regarding Blastocystis biology within and outside the host is limited. Analysis of the metabolites produced by this anaerobe could provide insights that can help map its metabolism and determine its role in both health and disease. Due to its controversial pathogenicity, these metabolites could define its deterministic role in microbiome's "health" and/or subsequently resolve Blastocystis' potential impact in gastrointestinal health. A common method for elucidating the presence of these metabolites is through 1H nuclear magnetic resonance (NMR). However, there are currently no described benchmarked methods available to extract metabolites from Blastocystis for 1H NMR analysis. Herein, several extraction solvents, lysis methods and incubation temperatures were compared for their usefulness as an extraction protocol for this protozoan. Following extraction, the samples were freeze-dried, re-solubilized and analysed with 1H NMR. The results demonstrate that carrying out the procedure at room temperature using methanol as an extraction solvent and bead bashing as a lysis technique provides a consistent, reproducible and efficient method to extract metabolites from Blastocystis for NMR.
Collapse
Affiliation(s)
- Jamie M. Newton
- Laboratory of Molecular & Evolutionary Parasitology, RAPID Group, School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK; (J.M.N.); (E.L.B.); (L.Y.)
- Wellcome Trust Biomolecular NMR Facility, School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK;
| | - Emma L. Betts
- Laboratory of Molecular & Evolutionary Parasitology, RAPID Group, School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK; (J.M.N.); (E.L.B.); (L.Y.)
| | - Lyto Yiangou
- Laboratory of Molecular & Evolutionary Parasitology, RAPID Group, School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK; (J.M.N.); (E.L.B.); (L.Y.)
| | - Jose Ortega Roldan
- Wellcome Trust Biomolecular NMR Facility, School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK;
| | - Anastasios D. Tsaousis
- Laboratory of Molecular & Evolutionary Parasitology, RAPID Group, School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK; (J.M.N.); (E.L.B.); (L.Y.)
- Correspondence: (A.D.T.); (G.S.T.)
| | - Gary S. Thompson
- Wellcome Trust Biomolecular NMR Facility, School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK;
- Correspondence: (A.D.T.); (G.S.T.)
| |
Collapse
|
33
|
Pace RM, Williams JE, Robertson B, Lackey KA, Meehan CL, Price WJ, Foster JA, Sellen DW, Kamau-Mbuthia EW, Kamundia EW, Mbugua S, Moore SE, Prentice AM, Kita DG, Kvist LJ, Otoo GE, Ruiz L, Rodríguez JM, Pareja RG, McGuire MA, Bode L, McGuire MK. Variation in Human Milk Composition Is Related to Differences in Milk and Infant Fecal Microbial Communities. Microorganisms 2021; 9:1153. [PMID: 34072117 PMCID: PMC8230061 DOI: 10.3390/microorganisms9061153] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 05/21/2021] [Accepted: 05/24/2021] [Indexed: 12/11/2022] Open
Abstract
Previously published data from our group and others demonstrate that human milk oligosaccharide (HMOs), as well as milk and infant fecal microbial profiles, vary by geography. However, little is known about the geographical variation of other milk-borne factors, such as lactose and protein, as well as the associations among these factors and microbial community structures in milk and infant feces. Here, we characterized and contrasted concentrations of milk-borne lactose, protein, and HMOs, and examined their associations with milk and infant fecal microbiomes in samples collected in 11 geographically diverse sites. Although geographical site was strongly associated with milk and infant fecal microbiomes, both sample types assorted into a smaller number of community state types based on shared microbial profiles. Similar to HMOs, concentrations of lactose and protein also varied by geography. Concentrations of HMOs, lactose, and protein were associated with differences in the microbial community structures of milk and infant feces and in the abundance of specific taxa. Taken together, these data suggest that the composition of human milk, even when produced by relatively healthy women, differs based on geographical boundaries and that concentrations of HMOs, lactose, and protein in milk are related to variation in milk and infant fecal microbial communities.
Collapse
Affiliation(s)
- Ryan M. Pace
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID 83844, USA;
| | - Janet E. Williams
- Department of Animal, Veterinary and Food Sciences, University of Idaho, Moscow, ID 83844, USA; (J.E.W.); (M.A.M.)
| | - Bianca Robertson
- Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence, Univeristy of California San Diego, La Jolla, CA 92093, USA; (B.R.); (L.B.)
- Department of Pediatrics, Univeristy of California San Diego, La Jolla, CA 92093, USA
| | - Kimberly A. Lackey
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID 83844, USA;
| | - Courtney L. Meehan
- Department of Anthropology, Washington State University, Pullman, WA 99164, USA;
| | - William J. Price
- Statistical Programs, College of Agricultural and Life Sciences, University of Idaho, Moscow, ID 83844, USA;
| | - James A. Foster
- Department of Biological Sciences, University of Idaho, Moscow, ID 83844, USA;
| | - Daniel W. Sellen
- Department of Anthropology, University of Toronto, Toronto, ON M5S 1A8, Canada;
| | | | - Egidioh W. Kamundia
- Department of Human Nutrition, Egerton University, Nakuru 20115, Kenya; (E.W.K.-M.); (E.W.K.); (S.M.)
| | - Samwel Mbugua
- Department of Human Nutrition, Egerton University, Nakuru 20115, Kenya; (E.W.K.-M.); (E.W.K.); (S.M.)
| | - Sophie E. Moore
- Department of Women and Children’s Health, King’s College London, London WC2R 2LS, UK;
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara P.O. Box 273, Gambia;
| | - Andrew M. Prentice
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara P.O. Box 273, Gambia;
| | - Debela G. Kita
- Department of Anthropology, Hawassa University, Hawassa P.O. Box 27601, Ethiopia;
| | - Linda J. Kvist
- Faculty of Medicine, Lund University, 221 00 Lund, Sweden;
| | - Gloria E. Otoo
- Department of Nutrition and Food Science, University of Ghana, Accra 00233, Ghana;
| | - Lorena Ruiz
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain;
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Juan M. Rodríguez
- Department of Nutrition and Food Science, Complutense University of Madrid, 28040 Madrid, Spain;
| | | | - Mark A. McGuire
- Department of Animal, Veterinary and Food Sciences, University of Idaho, Moscow, ID 83844, USA; (J.E.W.); (M.A.M.)
| | - Lars Bode
- Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence, Univeristy of California San Diego, La Jolla, CA 92093, USA; (B.R.); (L.B.)
- Department of Pediatrics, Univeristy of California San Diego, La Jolla, CA 92093, USA
| | - Michelle K. McGuire
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID 83844, USA;
| |
Collapse
|
34
|
Rousseaux A, Brosseau C, Le Gall S, Piloquet H, Barbarot S, Bodinier M. Human Milk Oligosaccharides: Their Effects on the Host and Their Potential as Therapeutic Agents. Front Immunol 2021; 12:680911. [PMID: 34108974 PMCID: PMC8180913 DOI: 10.3389/fimmu.2021.680911] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 04/29/2021] [Indexed: 12/15/2022] Open
Abstract
Breastmilk is known to be very important for infants because it provides nutrients and immunological compounds. Among these compounds, human milk oligosaccharides (HMOs) represent the third most important component of breastmilk after lipids and lactose. Several experiments demonstrated the beneficial effects of these components on the microbiota, the immune system and epithelial barriers, which are three major biological systems. Indeed, HMOs induce bacterial colonization in the intestinal tract, which is beneficial for health. The gut bacteria can act directly and indirectly on the immune system by stimulating innate immunity and controlling inflammatory reactions and by inducing an adaptive immune response and a tolerogenic environment. In parallel, HMOs directly strengthen the intestinal epithelial barrier, protecting the host against pathogens. Here, we review the molecular mechanisms of HMOs in these different compartments and highlight their potential use as new therapeutic agents, especially in allergy prevention.
Collapse
Affiliation(s)
- Anaïs Rousseaux
- INRAE, Biopolyméres Interactions Assemblages, Nantes, France
| | - Carole Brosseau
- INRAE, Biopolyméres Interactions Assemblages, Nantes, France
| | - Sophie Le Gall
- INRAE, Biopolyméres Interactions Assemblages, Nantes, France.,INRAE, Bioressources: Imagerie, Biochimie & Structure, Nantes, France
| | - Hugues Piloquet
- Centre Hospitalier Universitaire Nantes, UMR1280 PhAN, Nantes, France
| | | | - Marie Bodinier
- INRAE, Biopolyméres Interactions Assemblages, Nantes, France
| |
Collapse
|
35
|
Moubareck CA. Human Milk Microbiota and Oligosaccharides: A Glimpse into Benefits, Diversity, and Correlations. Nutrients 2021; 13:1123. [PMID: 33805503 PMCID: PMC8067037 DOI: 10.3390/nu13041123] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/18/2021] [Accepted: 03/26/2021] [Indexed: 12/12/2022] Open
Abstract
Human milk represents a cornerstone for growth and development of infants, with extensive array of benefits. In addition to exceptionally nutritive and bioactive components, human milk encompasses a complex community of signature bacteria that helps establish infant gut microbiota, contributes to maturation of infant immune system, and competitively interferes with pathogens. Among bioactive constituents of milk, human milk oligosaccharides (HMOs) are particularly significant. These are non-digestible carbohydrates forming the third largest solid component in human milk. Valuable effects of HMOs include shaping intestinal microbiota, imparting antimicrobial effects, developing intestinal barrier, and modulating immune response. Moreover, recent investigations suggest correlations between HMOs and milk microbiota, with complex links possibly existing with environmental factors, genetics, geographical location, and other factors. In this review, and from a physiological and health implications perspective, milk benefits for newborns and mothers are highlighted. From a microbiological perspective, a focused insight into milk microbiota, including origins, diversity, benefits, and effect of maternal diet is presented. From a metabolic perspective, biochemical, physiological, and genetic significance of HMOs, and their probable relations to milk microbiota, are addressed. Ongoing research into mechanistic processes through which the rich biological assets of milk promote development, shaping of microbiota, and immunity is tackled.
Collapse
Affiliation(s)
- Carole Ayoub Moubareck
- College of Natural and Health Sciences, Zayed University, Dubai 19282, United Arab Emirates
| |
Collapse
|
36
|
Boudry G, Charton E, Le Huerou-Luron I, Ferret-Bernard S, Le Gall S, Even S, Blat S. The Relationship Between Breast Milk Components and the Infant Gut Microbiota. Front Nutr 2021; 8:629740. [PMID: 33829032 PMCID: PMC8019723 DOI: 10.3389/fnut.2021.629740] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 02/16/2021] [Indexed: 12/12/2022] Open
Abstract
The assembly of the newborn's gut microbiota during the first months of life is an orchestrated process resulting in specialized microbial ecosystems in the different gut compartments. This process is highly dependent upon environmental factors, and many evidences suggest that early bacterial gut colonization has long-term consequences on host digestive and immune homeostasis but also metabolism and behavior. The early life period is therefore a "window of opportunity" to program health through microbiota modulation. However, the implementation of this promising strategy requires an in-depth understanding of the mechanisms governing gut microbiota assembly. Breastfeeding has been associated with a healthy microbiota in infants. Human milk is a complex food matrix, with numerous components that potentially influence the infant microbiota composition, either by enhancing specific bacteria growth or by limiting the growth of others. The objective of this review is to describe human milk composition and to discuss the established or purported roles of human milk components upon gut microbiota establishment. Finally, the impact of maternal diet on human milk composition is reviewed to assess how maternal diet could be a simple and efficient approach to shape the infant gut microbiota.
Collapse
Affiliation(s)
- Gaëlle Boudry
- Institut NuMeCan, INRAE, INSERM, Univ Rennes, Saint-Gilles, France
| | - Elise Charton
- Institut NuMeCan, INRAE, INSERM, Univ Rennes, Saint-Gilles, France
- UMR STLO INRAE, Institut Agro, Rennes, France
| | | | | | - Sophie Le Gall
- INRAE, UR BIA, Nantes, France
- INRAE, BIBS facility, Nantes, France
| | | | - Sophie Blat
- Institut NuMeCan, INRAE, INSERM, Univ Rennes, Saint-Gilles, France
| |
Collapse
|
37
|
Sindi AS, Geddes DT, Wlodek ME, Muhlhausler BS, Payne MS, Stinson LF. Can we modulate the breastfed infant gut microbiota through maternal diet? FEMS Microbiol Rev 2021; 45:6133472. [PMID: 33571360 DOI: 10.1093/femsre/fuab011] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/09/2021] [Indexed: 12/11/2022] Open
Abstract
Initial colonisation of the infant gut is robustly influenced by regular ingestion of human milk, a substance that contains microbes, microbial metabolites, immune proteins, and oligosaccharides. Numerous factors have been identified as potential determinants of the human milk and infant gut microbiota, including maternal diet; however, there is limited data on the influence of maternal diet during lactation on either of these. Here, we review the processes thought to contribute to human milk and infant gut bacterial colonisation and provide a basis for considering the role of maternal dietary patterns during lactation in shaping infant gut microbial composition and function. Although only one observational study has directly investigated the influence of maternal diet during lactation on the infant gut microbiome, data from animal studies suggests that modulation of the maternal gut microbiota, via diet or probiotics, may influence the mammary or milk microbiota. Additionally, evidence from human studies suggests that the maternal diet during pregnancy may affect the gut microbiota of the breastfed infant. Together, there is a plausible hypothesis that maternal diet during lactation may influence the infant gut microbiota. If substantiated in further studies, this may present a potential window of opportunity for modulating the infant gut microbiome in early life.
Collapse
Affiliation(s)
- Azhar S Sindi
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia.,College of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Donna T Geddes
- School of Molecular Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Mary E Wlodek
- Department of Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Beverly S Muhlhausler
- CSIRO, Adelaide, South Australia, Australia.,School of Agriculture, Food and Wine, The University of Adelaide, Adelaide, South Australia, Australia
| | - Matthew S Payne
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia
| | - Lisa F Stinson
- School of Molecular Sciences, The University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
38
|
The Metabolomic Analysis of Human Milk Offers Unique Insights into Potential Child Health Benefits. Curr Nutr Rep 2021; 10:12-29. [PMID: 33555534 DOI: 10.1007/s13668-020-00345-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2020] [Indexed: 10/22/2022]
Abstract
PURPOSE OF REVIEW Human milk is the gold standard of infant nutrition. The milk changes throughout lactation and is tailored for the infant providing the nutrients, minerals and vitamins necessary for supporting healthy infant growth. Human milk also contains low molecular weight compounds (metabolites) possibly eliciting important bioactivity. Metabolomics is the study of these metabolites. The purpose of this review was to examine recent metabolomics studies and cohort studies on human milk to assess the impact of human milk metabolomic analyses combined with investigations of infant growth and development. RECENT FINDINGS The metabolite profile of human milk varies among other factors according to lactation stage, gestation at birth, and maternal genes, diet and disease state. Few studies investigate how these variations impact infant growth and development. Several time-related factors affecting human milk metabolome are potentially ubiquitous among mothers, although maternal-related factors are heavily confounded, which complicates studies of metabolite abundancies and variabilities and further possibilities of observing cause and effect in infants.
Collapse
|
39
|
Neonatal intensive care unit (NICU) exposures exert a sustained influence on the progression of gut microbiota and metabolome in the first year of life. Sci Rep 2021; 11:1353. [PMID: 33446779 PMCID: PMC7809424 DOI: 10.1038/s41598-020-80278-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 12/18/2020] [Indexed: 12/19/2022] Open
Abstract
Emerging evidence has shown a link between the perturbations and development of the gut microbiota in infants with their immediate and long-term health. To better understand the assembly of the gut microbiota in preterm infants, faecal samples were longitudinally collected from the preterm (n = 19) and term (n = 20) infants from birth until month 12. 16S rRNA gene sequencing (n = 141) and metabolomics profiling (n = 141) using nuclear magnetic resonance spectroscopy identified significant differences between groups in various time points. A panel of amino acid metabolites and central metabolism intermediates significantly correlated with the relative abundances of 8 species of bacteria were identified in the preterm group. In contrast, faecal metabolites of term infants had significantly higher levels of metabolites which are commonly found in milk such as fucose and β-hydroxybutyrate. We demonstrated that the early-life factors such as gestational age, birth weight and NICU exposures, exerted a sustained effect to the dynamics of gut microbial composition and metabolism of the neonates up to one year of age. Thus, our findings suggest that intervention at this early time could provide ‘metabolic rescue’ to preterm infants from aberrant initial gut microbial colonisation and succession.
Collapse
|
40
|
Neonatal intensive care unit (NICU) exposures exert a sustained influence on the progression of gut microbiota and metabolome in the first year of life. Sci Rep 2021; 11:1353. [PMID: 33446779 PMCID: PMC7809424 DOI: 10.1038/s41598-020-80278-1 10.1038/s41598-021-88758-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
Abstract
Emerging evidence has shown a link between the perturbations and development of the gut microbiota in infants with their immediate and long-term health. To better understand the assembly of the gut microbiota in preterm infants, faecal samples were longitudinally collected from the preterm (n = 19) and term (n = 20) infants from birth until month 12. 16S rRNA gene sequencing (n = 141) and metabolomics profiling (n = 141) using nuclear magnetic resonance spectroscopy identified significant differences between groups in various time points. A panel of amino acid metabolites and central metabolism intermediates significantly correlated with the relative abundances of 8 species of bacteria were identified in the preterm group. In contrast, faecal metabolites of term infants had significantly higher levels of metabolites which are commonly found in milk such as fucose and β-hydroxybutyrate. We demonstrated that the early-life factors such as gestational age, birth weight and NICU exposures, exerted a sustained effect to the dynamics of gut microbial composition and metabolism of the neonates up to one year of age. Thus, our findings suggest that intervention at this early time could provide 'metabolic rescue' to preterm infants from aberrant initial gut microbial colonisation and succession.
Collapse
|
41
|
Lopez Leyva L, Brereton NJ, Koski KG. Emerging frontiers in human milk microbiome research and suggested primers for 16S rRNA gene analysis. Comput Struct Biotechnol J 2020; 19:121-133. [PMID: 33425245 PMCID: PMC7770459 DOI: 10.1016/j.csbj.2020.11.057] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/24/2020] [Accepted: 11/28/2020] [Indexed: 02/06/2023] Open
Abstract
Human milk is the ideal food for infants due to its unique nutritional and immune properties, and more recently human milk has also been recognized as an important source of bacteria for infants. However, a substantial amount of fundamental human milk microbiome information remains unclear, such as the origin, composition and function of the community and its members. There is emerging evidence to suggest that the diversity and composition of the milk microbiome might differ between lactation stages, due to maternal factors and diet, agrarian and urban lifestyles, and geographical location. The evolution of the methods used for studying milk microbiota, transitioning from culture dependent-approaches to include culture-independent approaches, has had an impact on findings and, in particular, primer selection within 16S rRNA gene barcoding studies have led to discrepancies in observed microbiota communities. Here, the current state-of-the-art is reviewed and emerging frontiers essential to improving our understanding of the human milk microbiome are considered.
Collapse
Affiliation(s)
- Lilian Lopez Leyva
- School of Human Nutrition, McGill University, Macdonald Stewart Building, 21111 Lakeshore Road, Ste-Anne de Bellevue, QC H9X 3V9, Canada
| | - Nicholas J.B. Brereton
- Institut de Recherche en Biologie Végétale, Université de Montréal, 4101 Sherbrooke St E, Montreal, QC H1X 2B2, Canada
| | - Kristine G. Koski
- School of Human Nutrition, McGill University, Macdonald Stewart Building, 21111 Lakeshore Road, Ste-Anne de Bellevue, QC H9X 3V9, Canada
| |
Collapse
|
42
|
Fernández L, Pannaraj PS, Rautava S, Rodríguez JM. The Microbiota of the Human Mammary Ecosystem. Front Cell Infect Microbiol 2020; 10:586667. [PMID: 33330129 PMCID: PMC7718026 DOI: 10.3389/fcimb.2020.586667] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/15/2020] [Indexed: 12/12/2022] Open
Abstract
Human milk contains a dynamic and complex site-specific microbiome, which is not assembled in an aleatory way, formed by organized microbial consortia and networks. Presence of some genera, such as Staphylococcus, Streptococcus, Corynebacterium, Cutibacterium (formerly known as Propionibacterium), Lactobacillus, Lactococcus and Bifidobacterium, has been detected by both culture-dependent and culture-independent approaches. DNA from some gut-associated strict anaerobes has also been repeatedly found and some studies have revealed the presence of cells and/or nucleic acids from viruses, archaea, fungi and protozoa in human milk. Colostrum and milk microbes are transmitted to the infant and, therefore, they are among the first colonizers of the human gut. Still, the significance of human milk microbes in infant gut colonization remains an open question. Clinical studies trying to elucidate the question are confounded by the profound impact of non-microbial human milk components to intestinal microecology. Modifications in the microbiota of human milk may have biological consequences for infant colonization, metabolism, immune and neuroendocrine development, and for mammary health. However, the factors driving differences in the composition of the human milk microbiome remain poorly known. In addition to colostrum and milk, breast tissue in lactating and non-lactating women may also contain a microbiota, with implications in the pathogenesis of breast cancer and in some of the adverse outcomes associated with breast implants. This and other open issues, such as the origin of the human milk microbiome, and the current limitations and future prospects are addressed in this review.
Collapse
Affiliation(s)
- Leónides Fernández
- Department of Galenic Pharmacy and Food Technology, Complutense University of Madrid, Madrid, Spain
| | - Pia S. Pannaraj
- Department of Pediatrics and Molecular Microbiology and Immunology, Keck School of Medicine and Children’s Hospital, Los Angeles, CA, United States
| | - Samuli Rautava
- University of Helsinki and Helsinki University Hospital, New Children’s Hospital, Pediatric Research Center, Helsinki, Finland
| | - Juan M. Rodríguez
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
43
|
Cutignano A, Siano F, Romano R, Aiello A, Pizzolongo F, Berni Canani R, Paparo L, Nocerino R, Di Scala C, Addeo F, Picariello G. Short-term effects of dietary bovine milk on fatty acid composition of human milk: A preliminary multi-analytical study. J Chromatogr B Analyt Technol Biomed Life Sci 2020; 1154:122189. [PMID: 32861173 DOI: 10.1016/j.jchromb.2020.122189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 05/13/2020] [Accepted: 05/20/2020] [Indexed: 12/11/2022]
Abstract
The fatty acid (FA) composition of human milk (HM) from N = 9 Italian healthy donors following a free diet exhibited FA-dependent ranges of variability, as assessed by GC-FID. The possible short-term changes in the FA profile were monitored in the milk of lactating mothers (three) collected at five time points over a 6 h period, following an oral load (200 mL) of bovine milk. An array of techniques was exploited, including UHPLC-ESI-MS/MS of intact lipids and MALDI-TOF MS before and after chemical hydrogenation or bromination, in addition to MALDI-TOF MS analysis of FA after saponification, to monitor short-chain and odd-chain FA in HM as markers of bovine milk fat. A single administration of bovine milk did not appreciably modify the lipid pattern, suggesting that the maternal diet could induce not detectable short-term changes on the lipid composition of HM. Diet-induced increase of butyric acid was also excluded by 13C NMR. The functions that HM FA exert in infant physiology appear finely regulated through maternal metabolism.
Collapse
Affiliation(s)
- Adele Cutignano
- Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche (CNR), Via Campi Flegrei 34, 80078 Pozzuoli (Napoli), Italy
| | - Francesco Siano
- Istituto di Scienze dell'Alimentazione, Consiglio Nazionale delle Ricerche (CNR), Via Roma 64, 83100 Avellino, Italy
| | - Raffaele Romano
- Dipartimento di Agraria, Università di Napoli "Federico II", Parco Gussone, 80055 Portici (Napoli), Italy
| | - Alessandra Aiello
- Dipartimento di Agraria, Università di Napoli "Federico II", Parco Gussone, 80055 Portici (Napoli), Italy
| | - Fabiana Pizzolongo
- Dipartimento di Agraria, Università di Napoli "Federico II", Parco Gussone, 80055 Portici (Napoli), Italy
| | - Roberto Berni Canani
- Dipartimento di Science Mediche Traslazionali e Laboratorio Europeo per lo Studio delle Malattie Indotte da Alimenti, Università degli Studi di Napoli Federico II, Via S. Pansini 5, 80131 Napoli, Italy; ImmunoNutritionLab at CEINGE Biotechinogie Avanzate, Università degli Studi di Napoli Federico II, Via Comunale Margherita, 484-538, 80131 Napoli, Italy; Task Force di Ateneo per gli Studi sul Microbioma, Università degli Studi di Napoli Federico II, Via S. Pansini 5, 80131 Napoli, Italy
| | - Lorella Paparo
- Dipartimento di Science Mediche Traslazionali e Laboratorio Europeo per lo Studio delle Malattie Indotte da Alimenti, Università degli Studi di Napoli Federico II, Via S. Pansini 5, 80131 Napoli, Italy; ImmunoNutritionLab at CEINGE Biotechinogie Avanzate, Università degli Studi di Napoli Federico II, Via Comunale Margherita, 484-538, 80131 Napoli, Italy; Task Force di Ateneo per gli Studi sul Microbioma, Università degli Studi di Napoli Federico II, Via S. Pansini 5, 80131 Napoli, Italy
| | - Rita Nocerino
- Dipartimento di Science Mediche Traslazionali e Laboratorio Europeo per lo Studio delle Malattie Indotte da Alimenti, Università degli Studi di Napoli Federico II, Via S. Pansini 5, 80131 Napoli, Italy; ImmunoNutritionLab at CEINGE Biotechinogie Avanzate, Università degli Studi di Napoli Federico II, Via Comunale Margherita, 484-538, 80131 Napoli, Italy; Task Force di Ateneo per gli Studi sul Microbioma, Università degli Studi di Napoli Federico II, Via S. Pansini 5, 80131 Napoli, Italy
| | - Carmen Di Scala
- Dipartimento di Science Mediche Traslazionali e Laboratorio Europeo per lo Studio delle Malattie Indotte da Alimenti, Università degli Studi di Napoli Federico II, Via S. Pansini 5, 80131 Napoli, Italy; ImmunoNutritionLab at CEINGE Biotechinogie Avanzate, Università degli Studi di Napoli Federico II, Via Comunale Margherita, 484-538, 80131 Napoli, Italy; Task Force di Ateneo per gli Studi sul Microbioma, Università degli Studi di Napoli Federico II, Via S. Pansini 5, 80131 Napoli, Italy
| | - Francesco Addeo
- Dipartimento di Agraria, Università di Napoli "Federico II", Parco Gussone, 80055 Portici (Napoli), Italy
| | - Gianluca Picariello
- Istituto di Scienze dell'Alimentazione, Consiglio Nazionale delle Ricerche (CNR), Via Roma 64, 83100 Avellino, Italy.
| |
Collapse
|
44
|
Quin C, Gibson DL. Human behavior, not race or geography, is the strongest predictor of microbial succession in the gut bacteriome of infants. Gut Microbes 2020; 11:1143-1171. [PMID: 32249675 PMCID: PMC7524360 DOI: 10.1080/19490976.2020.1736973] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Colonization of the gastrointestinal tract with microorganisms during infancy represents a critical control point for shaping life-long immune-mediated disease susceptibility. Abnormal colonization or an imbalance of microbes, termed dysbiosis, is implicated in several diseases. Consequently, recent research has aimed at understanding ways to manipulate a dysbiotic microbiome during infancy to resemble a normal, healthy microbiome. However, one of the fundamental issues in microbiome research is characterizing what a "normal" infant microbiome is based on geography, ethnicity and cultural variations. This review provides a comprehensive account of what is currently known about the infant microbiome from a global context. In general, this review shows that the influence of cultural variations in feeding practices, delivery modes and hygiene are the biggest contributors to microbial variability. Despite geography or race, all humans have similar microbial succession during infancy.
Collapse
Affiliation(s)
- Candice Quin
- Department of Biology, University of British Columbia, Kelowna, Canada
| | - Deanna L. Gibson
- Department of Biology, University of British Columbia, Kelowna, Canada,Department of Medicine, University of British Columbia, Kelowna, Canada,CONTACT Deanna L. Gibson Department of Biology, University of British Columbia, Okanagan Campus, ASC 386, 3187 University Way, Kelowna, BCV1V 1V7, Canada
| |
Collapse
|
45
|
Xu J, Lawley B, Wong G, Otal A, Chen L, Ying TJ, Lin X, Pang WW, Yap F, Chong YS, Gluckman PD, Lee YS, Chong MFF, Tannock GW, Karnani N. Ethnic diversity in infant gut microbiota is apparent before the introduction of complementary diets. Gut Microbes 2020; 11:1362-1373. [PMID: 32453615 PMCID: PMC7524347 DOI: 10.1080/19490976.2020.1756150] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The human gut microbiota develops soon after birth and can acquire inter-individual variation upon exposure to intrinsic and environmental cues. However, inter-individual variation has not been comprehensively assessed in a multi-ethnic study. We studied a longitudinal birth cohort of 106 infants of three Asian ethnicities (Chinese, Malay, and Indian) that resided in the same geographical location (Singapore). Specific and temporal influences of ethnicity, mode of delivery, breastfeeding status, gestational age, birthweight, gender, and maternal education on the development of the gut microbiota in the first 2 years of life were studied. Mode of delivery, breastfeeding status, and ethnicity were identified as the main factors influencing the compositional development of the gut microbiota. Effects of delivery mode and breastfeeding status lasted until 6M and 3M, respectively, with the primary impact on the diversity and temporal colonization of the genera Bacteroides and Bifidobacterium. The effect of ethnicity was apparent at 3M post-birth, even before the introduction of weaning (complementary) foods, and remained significant after adjusting for delivery mode and breastfeeding status. Ethnic influences remained significant until 12M in the Indian and Chinese infants. The microbiota of Indian infants was characterized by higher abundances of Bifidobacterium and Lactobacillus, while Chinese infants had higher abundances of Bacteroides and Akkermansia. These findings provide a detailed insight into the specific and temporal influences of early life factors and ethnicity in the development of the human gut microbiota. Trial Registration: Clinicaltrials.gov registration no. NCT01174875.
Collapse
Affiliation(s)
- Jia Xu
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore
| | - Blair Lawley
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Gerard Wong
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore
| | - Anna Otal
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Li Chen
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore
| | - Toh Jia Ying
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore
| | - Xinyi Lin
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore,Centre for Quantitative Medicine and Program in Health Services and Systems Research, Duke-NUS Medical School, Singapore
| | - Wei Wei Pang
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Fabian Yap
- Department of Pediatric Endocrinology, KK Women’s and Children’s Hospital, Singapore
| | - Yap-Seng Chong
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore,Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Peter D. Gluckman
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore,Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Yung Seng Lee
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore,Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore,Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Mary Foong-Fong Chong
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore,Clinical Nutrition Research Centre, Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore,Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | - Gerald W. Tannock
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand,Gerald W. Tannock Department of Microbiology and Immunology, University of Otago, Dunedin9054, New Zealand
| | - Neerja Karnani
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore,CONTACT Neerja Karnani Brenner Centre for Molecular Medicine, Singapore Institute for Clinical Sciences (SICS), A*STAR, 30 Medical Drive117609, Singapore
| |
Collapse
|
46
|
Stinson LF, Gay MCL, Koleva PT, Eggesbø M, Johnson CC, Wegienka G, du Toit E, Shimojo N, Munblit D, Campbell DE, Prescott SL, Geddes DT, Kozyrskyj AL. Human Milk From Atopic Mothers Has Lower Levels of Short Chain Fatty Acids. Front Immunol 2020; 11:1427. [PMID: 32903327 PMCID: PMC7396598 DOI: 10.3389/fimmu.2020.01427] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 06/03/2020] [Indexed: 12/16/2022] Open
Abstract
Short chain fatty acids (SFCAs) are microbial metabolites produced in the gut upon fermentation of dietary fiber. These metabolites interact with the host immune system and can elicit epigenetic effects. There is evidence to suggest that SCFAs may play a role in the developmental programming of immune disorders and obesity, though evidence in humans remains sparse. Here we have quantified human milk (HM) SCFA levels in an international cohort of atopic and non-atopic mothers (n = 109). Our results demonstrate that human milk contains detectable levels of the SCFAs acetate, butyrate, and formate. Samples from atopic mothers had significantly lower concentrations of acetate and butyrate than those of non-atopic mothers. HM SCFA levels in atopic and non-atopic women also varied based on maternal country of residence (Australia, Japan, Norway, South Africa, USA). Reduced exposure to HM SCFA in early life may program atopy or overweight risk in breastfed infants.
Collapse
Affiliation(s)
- Lisa F Stinson
- School of Molecular Sciences, University of Western Australia, Perth, WA, Australia.,inVIVO Planetary Health of the Worldwide Universities Network (WUN), West New York, NJ, United States
| | - Melvin C L Gay
- School of Molecular Sciences, University of Western Australia, Perth, WA, Australia.,inVIVO Planetary Health of the Worldwide Universities Network (WUN), West New York, NJ, United States
| | - Petya T Koleva
- inVIVO Planetary Health of the Worldwide Universities Network (WUN), West New York, NJ, United States.,Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Merete Eggesbø
- inVIVO Planetary Health of the Worldwide Universities Network (WUN), West New York, NJ, United States.,Department of Environmental Exposure and Epidemiology, Norwegian Institute of Public Health, Oslo, Norway
| | - Christine C Johnson
- inVIVO Planetary Health of the Worldwide Universities Network (WUN), West New York, NJ, United States.,Department of Public Health Sciences, Henry Ford Hospital, Detroit, MI, United States
| | - Ganesa Wegienka
- inVIVO Planetary Health of the Worldwide Universities Network (WUN), West New York, NJ, United States.,Department of Public Health Sciences, Henry Ford Hospital, Detroit, MI, United States
| | - Elloise du Toit
- inVIVO Planetary Health of the Worldwide Universities Network (WUN), West New York, NJ, United States.,Division of Medical Microbiology, University of Cape Town, Cape Town, South Africa
| | - Naoki Shimojo
- inVIVO Planetary Health of the Worldwide Universities Network (WUN), West New York, NJ, United States.,Department of Pediatrics, Chiba University, Chiba, Japan
| | - Daniel Munblit
- inVIVO Planetary Health of the Worldwide Universities Network (WUN), West New York, NJ, United States.,Department of Paediatrics and Paediatric Infectious Diseases, Institute of Child Health, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia.,Inflammation, Repair and Development Section, National Heart & Lung Institute, Imperial College London, London, United Kingdom
| | - Dianne E Campbell
- inVIVO Planetary Health of the Worldwide Universities Network (WUN), West New York, NJ, United States.,Department of Allergy and Immunology, Children's Hospital at Westmead, University of Sydney, Sydney, NSW, Australia
| | - Susan L Prescott
- inVIVO Planetary Health of the Worldwide Universities Network (WUN), West New York, NJ, United States.,The ORIGINS Project, Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| | - Donna T Geddes
- School of Molecular Sciences, University of Western Australia, Perth, WA, Australia.,inVIVO Planetary Health of the Worldwide Universities Network (WUN), West New York, NJ, United States
| | - Anita L Kozyrskyj
- inVIVO Planetary Health of the Worldwide Universities Network (WUN), West New York, NJ, United States.,Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
47
|
Salminen S, Stahl B, Vinderola G, Szajewska H. Infant Formula Supplemented with Biotics: Current Knowledge and Future Perspectives. Nutrients 2020; 12:E1952. [PMID: 32629970 PMCID: PMC7400136 DOI: 10.3390/nu12071952] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 06/27/2020] [Accepted: 06/28/2020] [Indexed: 02/06/2023] Open
Abstract
Breastfeeding is natural and the optimal basis of infant nutrition and development, with many benefits for maternal health. Human milk is a dynamic fluid fulfilling an infant's specific nutritional requirements and guiding the growth, developmental, and physiological processes of the infant. Human milk is considered unique in composition, and it is influenced by several factors, such as maternal diet and health, body composition, and geographic region. Human milk stands as a model for infant formula providing nutritional solutions for infants not able to receive enough mother's milk. Infant formulas aim to mimic the composition and functionality of human milk by providing ingredients reflecting those of the latest human milk insights, such as oligosaccharides, bacteria, and bacterial metabolites. The objective of this narrative review is to discuss the most recent developments in infant formula with a special focus on human milk oligosaccharides and postbiotics.
Collapse
Affiliation(s)
- Seppo Salminen
- Functional Foods Forum, Faculty of Medicine, University of Turku, 20520 Turku, Finland;
| | - Bernd Stahl
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands;
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Gabriel Vinderola
- Instituto de Lactología Industrial (INLAIN, UNL-CONICET), Facultad de Ingeniería Química, Universidad Nacional del Litoral, Santiago del Estero 2829, Santa Fe 3000, Argentina;
| | - Hania Szajewska
- Department of Paediatrics at the Medical University of Warsaw, 02091 Warsaw, Poland
| |
Collapse
|
48
|
Profiles of Human Milk Oligosaccharides and Their Relations to the Milk Microbiota of Breastfeeding Mothers in Dubai. Nutrients 2020; 12:nu12061727. [PMID: 32526930 PMCID: PMC7353065 DOI: 10.3390/nu12061727] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/27/2020] [Accepted: 06/03/2020] [Indexed: 12/12/2022] Open
Abstract
The composition of human breast milk is affected by several factors, including genetics, geographic location and maternal nutrition. This study investigated the human milk oligosaccharides (HMOs) of breastfeeding mothers living in Dubai and their relations with the milk microbiota. A total of 30 breast milk samples were collected from healthy Emirati and UAE-expatriates at Latifa Hospital. HMO profiling was performed using UHPLC-MS. Microbiota profiles were determined by sequencing amplicons of the V3-V4 region of the 16S rRNA gene. HMO concentrations were significantly higher in Emirati, and dropped with the lactation period in both groups of mothers. The Le (a-b+)-secretor (Le+Se+) type was the most abundant in Dubai mothers (60%), followed by the Le(a-b-)-secretor (Le-Se+) type (23%). Bifidobacterium and Lactobacillus were considerably lower in Dubai-based mothers, while Pseudomonas and Delftia (Hydrogenophaga) were detected at a higher abundance compared to mothers from other countries. Atopobium was correlated with sialyl-lacto-N-tetraose c, Leptotrichia and Veillonella were correlated with 6'-sialyl-lactose, and Porphyromonas was correlated with lacto-N-hexaose. The study highlights the HMO profiles of breastfeeding mothers in Dubai and reveals few correlations with milk microbial composition. Targeted genomic analyses may help in determining whether these differences are due to genetic variations or to sociocultural and environmental factors.
Collapse
|
49
|
Demmelmair H, Jiménez E, Collado MC, Salminen S, McGuire MK. Maternal and Perinatal Factors Associated with the Human Milk Microbiome. Curr Dev Nutr 2020; 4:nzaa027. [PMID: 32270132 PMCID: PMC7127925 DOI: 10.1093/cdn/nzaa027] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 02/12/2020] [Accepted: 02/26/2020] [Indexed: 12/13/2022] Open
Abstract
Microbes are present in human milk regardless of the mother's health. The origins of the milk microbiota likely include the mother's skin, infant's mouth, and transfer from the maternal gastrointestinal (GI) tract. Prominent bacterial taxa in human milk are Staphylococcus and Streptococcus, but many other genera are also found including anaerobic Lactobacillus, Bifidobacterium, and Bacteroides. The milk microbiome is highly variable and potentially influenced by geographic location, delivery mode, time postpartum, feeding mode, social networks, environment, maternal diet, and milk composition. Mastitis alters the milk microbiome, and the intake of Lactobacilli has shown potential for mastitis treatment and prevention. Although milk and infant fecal microbiomes are different, their variations appear to be related - suggesting that milk is an important contributor of early GI colonization. Nonetheless, nothing is known regarding whether the milk microbiome influences infant health. Further research and clinical interventions are needed to determine if changes in the microbiomes of human milk and infant formula/food impact health.
Collapse
Affiliation(s)
- Hans Demmelmair
- Dr. von Hauner Children´s Hospital, University of Munich Medical Center, Munich, Germany
| | - Esther Jiménez
- ProbiSearch SLU, Madrid, Spain
- Department of Nutrition, Food Science, and Technology, University Complutense, Madrid, Spain
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology, Spanish National Research Council, Valencia, Spain
- Functional Foods Forum, University of Turku, Turku, Finland
| | - Seppo Salminen
- Functional Foods Forum, University of Turku, Turku, Finland
| | - Michelle K McGuire
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID, USA
| |
Collapse
|
50
|
Nuclear Magnetic Resonance Metabolomics Reveals Qualitative and Quantitative Differences in the Composition of Human Breast Milk and Milk Formulas. Nutrients 2020; 12:nu12040921. [PMID: 32230787 PMCID: PMC7230615 DOI: 10.3390/nu12040921] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 01/21/2023] Open
Abstract
Commercial formula milk (FM) constitutes the best alternative to fulfill the nutritional requirements of infants when breastfeeding is precluded. Here, we present the comparative study of polar metabolite composition of human breast milk (HBM) and seven different brands of FM by nuclear magnetic resonance spectroscopy. The results of the multivariate data analysis exposed qualitative and quantitative differences between HBM and FM composition as well as within FM of various brands and in HBM itself (between individual mothers and lactation period). Several metabolites were found exclusively in HBM and FM. Statistically significant higher levels of isoleucine and methionine in their free form were detected in FM samples based on caprine milk, while FM samples based on bovine milk showed a higher level of glucose and galactose in comparison to HBM. The results suggest that the amelioration of FM formulation is imperative to better mimic the composition of minor nutrients in HBM.
Collapse
|