1
|
Asenova A, Hristova H, Ivanova S, Miteva V, Zhivkova I, Stefanova K, Moncheva P, Nedeva T, Urshev Z, Marinova-Yordanova V, Georgieva T, Tzenova M, Russinova M, Borisova T, Donchev D, Hristova P, Rasheva I. Identification and Characterization of Human Breast Milk and Infant Fecal Cultivable Lactobacilli Isolated in Bulgaria: A Pilot Study. Microorganisms 2024; 12:1839. [PMID: 39338513 PMCID: PMC11433867 DOI: 10.3390/microorganisms12091839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
During the last few decades, the main focus of numerous studies has been on the human breast milk microbiota and its influence on the infant intestinal microbiota and overall health. The presence of lactic acid bacteria in breast milk affects both the quantitative and qualitative composition of the infant gut microbiota. The aim of this study was to assess the most frequently detected cultivable rod-shaped lactobacilli, specific for breast milk of healthy Bulgarian women and fecal samples of their infants over the first month of life, in 14 mother-infant tandem pairs. Additionally, we evaluated the strain diversity among the most common isolated species. A total of 68 Gram-positive and catalase-negative strains were subjected to identification using the MALDI-TOF technique. Predominant cultivable populations belonging to the rod-shaped lactic acid bacteria have been identified as Lacticaseibacillus rhamnosus, Limosilactobacillus fermentum, Lacticaseibacillus paracasei, and Limosilactobacillus reuteri. Also, we confirmed the presence of Lactiplantibacillus plantarum and Lactobacillus gasseri. Up to 26 isolates were selected as representatives and analyzed by 16S rRNA sequencing for strain identity confirmation and a phylogenetic tree based on 16S rRNA gene sequence was constructed. Comparative analysis by four RAPD primers revealed genetic differences between newly isolated predominant L. rhamnosus strains. This pilot study provides data for the current first report concerning the investigation of the characteristic cultivable lactobacilli isolated from human breast milk and infant feces in Bulgaria.
Collapse
Affiliation(s)
- Asya Asenova
- Department of General and Industrial Microbiology, Faculty of Biology, Sofia University St. Kliment Ohridski, Dragan Tsankov Blvd 8, 1164 Sofia, Bulgaria; (A.A.); (T.N.); (P.H.)
| | - Hristiyana Hristova
- Department of General and Industrial Microbiology, Faculty of Biology, Sofia University St. Kliment Ohridski, Dragan Tsankov Blvd 8, 1164 Sofia, Bulgaria; (A.A.); (T.N.); (P.H.)
| | - Stanimira Ivanova
- Department of General and Industrial Microbiology, Faculty of Biology, Sofia University St. Kliment Ohridski, Dragan Tsankov Blvd 8, 1164 Sofia, Bulgaria; (A.A.); (T.N.); (P.H.)
| | - Viliana Miteva
- Department of General and Industrial Microbiology, Faculty of Biology, Sofia University St. Kliment Ohridski, Dragan Tsankov Blvd 8, 1164 Sofia, Bulgaria; (A.A.); (T.N.); (P.H.)
| | - Ivelina Zhivkova
- Department of Clinical Microbiology, National Center of Infectious and Parasitic Disease, Yanko Sakuzov Blvd 26, 1504 Sofia, Bulgaria (D.D.)
| | - Katerina Stefanova
- Agrobioinstitute Bulgarian Agriculture Academy, Dragan Tsankov Blvd 8, 1164 Sofia, Bulgaria;
| | - Penka Moncheva
- Department of General and Industrial Microbiology, Faculty of Biology, Sofia University St. Kliment Ohridski, Dragan Tsankov Blvd 8, 1164 Sofia, Bulgaria; (A.A.); (T.N.); (P.H.)
| | - Trayana Nedeva
- Department of General and Industrial Microbiology, Faculty of Biology, Sofia University St. Kliment Ohridski, Dragan Tsankov Blvd 8, 1164 Sofia, Bulgaria; (A.A.); (T.N.); (P.H.)
| | - Zoltan Urshev
- LB Bulgaricum PLC, Malashevska Str. 14, 1113 Sofia, Bulgaria
| | - Victoria Marinova-Yordanova
- Department of General and Industrial Microbiology, Faculty of Biology, Sofia University St. Kliment Ohridski, Dragan Tsankov Blvd 8, 1164 Sofia, Bulgaria; (A.A.); (T.N.); (P.H.)
- Department of Bioactivity of Compouds, Centre of Competence “Sustainable Utilization of Bio Resources and Waste of Medicinal and Aromatic Plants for Innovative Bioactive Products”, Dragan Tsankov Blvd 8, 1164 Sofia, Bulgaria
| | - Tzveta Georgieva
- Department of Applied Genomics and GMO, National Center of Public Health and Analyses, Academic Ivan Geshov Blvd 15, 1431 Sofia, Bulgaria;
| | - Margarita Tzenova
- Department of General and Industrial Microbiology, Faculty of Biology, Sofia University St. Kliment Ohridski, Dragan Tsankov Blvd 8, 1164 Sofia, Bulgaria; (A.A.); (T.N.); (P.H.)
| | - Maria Russinova
- Human Milk Bank, Sava Mihailov Str. 57, 1309 Sofia, Bulgaria (T.B.)
| | | | - Deyan Donchev
- Department of Clinical Microbiology, National Center of Infectious and Parasitic Disease, Yanko Sakuzov Blvd 26, 1504 Sofia, Bulgaria (D.D.)
- Centre of Competence “Fundamental Translational and Clinical Research in Infection and Immunity”, Yanko Sakuzov Blvd 26, 1504 Sofia, Bulgaria
| | - Petya Hristova
- Department of General and Industrial Microbiology, Faculty of Biology, Sofia University St. Kliment Ohridski, Dragan Tsankov Blvd 8, 1164 Sofia, Bulgaria; (A.A.); (T.N.); (P.H.)
| | - Iliyana Rasheva
- Department of General and Industrial Microbiology, Faculty of Biology, Sofia University St. Kliment Ohridski, Dragan Tsankov Blvd 8, 1164 Sofia, Bulgaria; (A.A.); (T.N.); (P.H.)
| |
Collapse
|
2
|
Dombrowska-Pali A, Wiktorczyk-Kapischke N, Chrustek A, Olszewska-Słonina D, Gospodarek-Komkowska E, Socha MW. Human Milk Microbiome-A Review of Scientific Reports. Nutrients 2024; 16:1420. [PMID: 38794658 PMCID: PMC11124344 DOI: 10.3390/nu16101420] [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: 04/05/2024] [Revised: 05/01/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
One of the most important bioactive components of breast milk are free breast milk oligosaccharides, which are a source of energy for commensal intestinal microorganisms, stimulating the growth of Bifidobacterium, Lactobacillus, and Bacteroides in a child's digestive tract. There is some evidence that maternal, perinatal, and environmental-cultural factors influence the modulation of the breast milk microbiome. This review summarizes research that has examined the composition of the breast milk microbiome and the factors that may influence it. The manuscript highlights the potential importance of the breast milk microbiome for the future development and health of children. The origin of bacteria in breast milk is thought to include the mother's digestive tract (entero-mammary tract), bacterial exposure to the breast during breastfeeding, and the retrograde flow of breast milk from the infant's mouth to the woman's milk ducts. Unfortunately, despite increasingly more precise methods for assessing microorganisms in human milk, the topic of the human milk microbiome is still quite limited and requires scientific research that takes into account various conditions.
Collapse
Affiliation(s)
- Agnieszka Dombrowska-Pali
- Department of Perinatology, Gynecology and Gynecologic Oncology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Łukasiewicza 1, 85-821 Bydgoszcz, Poland;
| | - Natalia Wiktorczyk-Kapischke
- Department of Microbiology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie-Skłodowskiej 9, 85-094 Bydgoszcz, Poland; (N.W.-K.); (E.G.-K.)
| | - Agnieszka Chrustek
- Department of Pathobiochemistry and Clinical Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie-Skłodowskiej 9, 85-094 Bydgoszcz, Poland; (A.C.); (D.O.-S.)
| | - Dorota Olszewska-Słonina
- Department of Pathobiochemistry and Clinical Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie-Skłodowskiej 9, 85-094 Bydgoszcz, Poland; (A.C.); (D.O.-S.)
| | - Eugenia Gospodarek-Komkowska
- Department of Microbiology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie-Skłodowskiej 9, 85-094 Bydgoszcz, Poland; (N.W.-K.); (E.G.-K.)
| | - Maciej W. Socha
- Department of Perinatology, Gynecology and Gynecologic Oncology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Łukasiewicza 1, 85-821 Bydgoszcz, Poland;
- Department of Obstetrics and Gynecology, St. Adalberts’s Hospital in Gdańsk, Copernicus Healthcare Entity LLC, Jana Pawła II 50, 80-462 Gdańsk, Poland
| |
Collapse
|
3
|
González A, Fullaondo A, Odriozola A. Impact of evolution on lifestyle in microbiome. ADVANCES IN GENETICS 2024; 111:149-198. [PMID: 38908899 DOI: 10.1016/bs.adgen.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/24/2024]
Abstract
This chapter analyses the interaction between microbiota and humans from an evolutionary point of view. Long-term interactions between gut microbiota and host have been generated as a result of dietary choices through coevolutionary processes, where mutuality of advantage is essential. Likewise, the characteristics of the intestinal environment have made it possible to describe different intrahost evolutionary mechanisms affecting microbiota. For its part, the intestinal microbiota has been of great importance in the evolution of mammals, allowing the diversification of dietary niches, phenotypic plasticity and the selection of host phenotypes. Although the origin of the human intestinal microbial community is still not known with certainty, mother-offspring transmission plays a key role, and it seems that transmissibility between individuals in adulthood also has important implications. Finally, it should be noted that certain aspects inherent to modern lifestyle, including refined diets, antibiotic intake, exposure to air pollutants, microplastics, and stress, could negatively affect the diversity and composition of our gut microbiota. This chapter aims to combine current knowledge to provide a comprehensive view of the interaction between microbiota and humans throughout evolution.
Collapse
Affiliation(s)
- Adriana González
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain.
| | - Asier Fullaondo
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Adrián Odriozola
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain
| |
Collapse
|
4
|
Filatava EJ, Liu Z, Xie J, Tran DB, Chen K, El Habbal N, Weinstock G, Zhou Y, Gregory KE. The preterm human milk microbiota fluctuates by postpartum week and is characterized by gestational age and maternal BMI. mBio 2023; 14:e0210623. [PMID: 37975676 PMCID: PMC10746270 DOI: 10.1128/mbio.02106-23] [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: 09/22/2023] [Accepted: 09/28/2023] [Indexed: 11/19/2023] Open
Abstract
IMPORTANCE Despite a growing recognition that the type of nutrition received by preterm infants influences their intestinal microbiome and health outcomes, the microbiota of mother's own milk (MOM), pasteurized donor human milk (PDHM), and infant formula remain poorly characterized. In our study, we found that the structure of microbial communities, bacterial diversity, and relative abundances of specific genera were significantly different between MOM, PDHM, and formula. Additionally, our results suggest that the microbiota of MOM changes as a function of time and maternal factors. Lastly, we identified three lactotypes within MOM that have distinct microbial compositions and described the maternal factors associated with them. These findings set the stage for future research aimed at advancing our knowledge of the microbiota of preterm infant nutrition and the specific influence it may have on health outcomes.
Collapse
Affiliation(s)
| | - Zhongmao Liu
- University of Connecticut, Storrs, Connecticut, USA
| | - Jiaojiao Xie
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | | | - Kun Chen
- University of Connecticut, Storrs, Connecticut, USA
| | | | | | - Yanjiao Zhou
- University of Connecticut, Storrs, Connecticut, USA
| | | |
Collapse
|
5
|
Jin X, Xiao J, Lu C, Ma W, Fan Y, Xue X, Xia Y, Chen N, Liu J, Pei X. Breastmilk microbiome changes associated with lactational mastitis and treatment with dandelion extract. Front Microbiol 2023; 14:1247868. [PMID: 38029215 PMCID: PMC10679338 DOI: 10.3389/fmicb.2023.1247868] [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: 06/26/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Dandelion (Pugongying) is one of the most frequently used Chinese herbs for treating lactational mastitis (LM). Pugongying granules, a patented medication primarily comprised of dandelion extract, have been approved by CFDA for LM treatment in China. The aims of this study were to investigate the etiology of LM and the mechanism by which Pugongying granules decrease LM symptoms, with a particular focus on the microbial communities found in breastmilk. Methods Participants were recruited from a previously performed randomized controlled trial (Identifier: NCT03756324, ClinicalTrials.gov). Between 2019 and 2020, women diagnosed with unilateral LM at the Beijing University of Chinese Medicine Third Affiliated Hospital were enrolled. In total, 42 paired breastmilk samples from the healthy and affected breasts of the participants were collected. Additionally, 37 paired pre- and post-treatment breastmilk samples from the affected breast were collected from women who received a 3-day course of either Pugongying granules (20 women) or cefdinir (17 women). Clinical outcomes [e.g., body temperature, visual analogue scale (VAS) score for breast pain, the percentage of neutrophils (NE%)] were analyzed pre- and post-treatment, and the breastmilk samples were subjected to 16S rRNA gene sequencing to analyze the alpha and beta diversities and identify significant bacteria. Finally, the relationship between microorganisms and clinical outcomes was analyzed. Results There was no significant difference in fever and pain between the Pugongying group and cefdinir group. The most prevalent bacterial genera in breastmilk were Streptococcus and Staphylococcus. Compared to healthy breastmilk, microbial diversity was reduced in affected breastmilk, and there was a higher relative abundance of Streptococcus. After Pugongying treatment, there was an increase in microbial diversity with significantly higher abundance of Corynebacterium. A negative correlation was found between Corynebacterium, VAS score, and NE%. Treatment with cefdinir did not affect microbial diversity. Taken together, our results show a correlation between LM and reduced microbial diversity, as well as an increased abundance of Streptococcus in affected breastmilk. Conclusion Pugongying granules enhanced microbial diversity in breastmilk samples. Given the substantial variation in individual microbiomes, identifying specific species of Streptococcus and Corynebacterium associated with LM may provide additional insight into LM pathogenesis and treatment.
Collapse
Affiliation(s)
- Xinyan Jin
- Centre for Evidence-based Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine Affiliated Xiamen Hospital, Xiamen, China
| | - Jinhe Xiao
- Department of Prevention and Treatment of Breast Disease, Haidian District Maternal and Child Health Care Hospital, Beijing, China
| | - Chunli Lu
- Guangdong Provincial Research Center of Integration of Traditional Chinese Medicine and Western Medicine in Metabolic Diseases (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, China
| | - Wenxin Ma
- Centre for Evidence-based Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yingyi Fan
- Department of Breast Surgery, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Xue Xue
- The First Clinical Medical School, Hubei University of Chinese Medicine, Wuhan, China
| | - Yaru Xia
- Department of Breast Surgery, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Nana Chen
- Department of Breast Surgery, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Jianping Liu
- Centre for Evidence-based Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaohua Pei
- Beijing University of Chinese Medicine Affiliated Xiamen Hospital, Xiamen, China
| |
Collapse
|
6
|
Yu R, Ma Y, Luo Z, Qi C, Xie A, Jiang Y, Zhu B, Sun J. Maternal supplementation with Limosilactobacillus reuteri FN041 for preventing infants with atopic dermatitis: study protocol for a randomized controlled trial. Front Microbiol 2023; 14:1267448. [PMID: 37869669 PMCID: PMC10588667 DOI: 10.3389/fmicb.2023.1267448] [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: 07/26/2023] [Accepted: 08/28/2023] [Indexed: 10/24/2023] Open
Abstract
Background Atopic dermatitis (AD) has increased rapidly with rapid urbanization; however, the treatment options for AD are lacking because the commonly used therapies can only alleviate symptoms. Limosilactobacillus reuteri (L. reuteri), FN041 is a specific strain isolated from human breast milk, and its protective potential against AD has been confirmed. This study aims to assess the efficacy of maternal consumption of L. reuteri FN041 during late pregnancy and lactation in preventing infantile AD. Methods First, a randomized, double-blind, placebo-controlled intervention study will be conducted on 340 pregnant females with babies at high risk for AD. These subjects will be randomly divided into four groups of different doses of L. reuteri FN041 (1 × 109, 5 × 109, and 1 × 1010 CFU/d) along with a placebo. The safety and efficacy of maternal use of L. reuteri FN041 for preventing infantile AD will be analyzed, and the most efficient dosage of L. reuteri FN041 will be determined. Subsequently, a multicenter cohort study of 500 pregnant females with babies at high risk for AD will be conducted to promote the maternal application of L. reuteri FN041. These subjects will be administered L. reuteri FN041 at the optimal dose determined during the first stage of late pregnancy and lactation, and their babies will be analyzed for AD development. Recruitment was initiated in October 2022. Discussion The primary outcome is the cumulative incidence of AD at 24 months after maternal consumption of L. reuteri FN041 during late pregnancy and lactation, whereas the secondary outcome is the efficiency of L. reuteri FN041 transfer from the mother's gut to breast milk and then the infant's gut after oral supplementation. This study will demonstrate the efficacy of edible probiotics isolated from breast milk in preventing or treating AD in infants. Accordingly, we provide population-based advice for administering specific probiotics for the primary prevention of AD in pregnant females. Understanding the underlying mechanisms of probiotic strains derived from breast milk can promote their application in preventing infant diseases associated with intestinal microbiota imbalance and immune disorders. Clinical trial registration https://www.chictr.org.cn/, identifier [ChiCTR2300075611].
Collapse
Affiliation(s)
- Renqiang Yu
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Women’s Hospital of Jiangnan University, Wuxi, China
| | - Yizhe Ma
- Department of Pediatric, Jiangyin People's Hospital of Nantong University, Wuxi, China
| | - Zichen Luo
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Women’s Hospital of Jiangnan University, Wuxi, China
| | - Ce Qi
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Anni Xie
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Women’s Hospital of Jiangnan University, Wuxi, China
| | - Yifan Jiang
- School of Medicine, Nantong University, Nantong, China
| | - Baoli Zhu
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Women’s Hospital of Jiangnan University, Wuxi, China
| | - Jin Sun
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| |
Collapse
|
7
|
Mandolo J, Parker EPK, Bronowski C, Sindhu KNC, Darby AC, Cunliffe NA, Kang G, Iturriza-Gómara M, Kamng’ona AW, Jere KC. Association Between Maternal Breastmilk Microbiota Composition and Rotavirus Vaccine Response in African, Asian, and European Infants: A Prospective Cohort Study. J Infect Dis 2023; 228:637-645. [PMID: 37364376 PMCID: PMC10469347 DOI: 10.1093/infdis/jiad234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/30/2023] [Accepted: 06/22/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND Maternal breastmilk is a source of pre- and pro-biotics that impact neonatal gut microbiota colonization. Because oral rotavirus vaccines (ORVs) are administered at a time when infants are often breastfed, breastmilk microbiota composition may have a direct or indirect influence on vaccine take and immunogenicity. METHODS Using standardized methods across sites, we compared breastmilk microbiota composition in relation to geographic location and ORV response in cohorts prospectively followed from birth to 18 weeks of age in India (n = 307), Malawi (n = 119), and the United Kingdom ([UK] n = 60). RESULTS Breastmilk microbiota diversity was higher in India and Malawi than the UK across 3 longitudinal samples spanning weeks of life 1 to 13. Dominant taxa such as Streptococcus and Staphylococcus were consistent across cohorts; however, significant geographic differences were observed in the prevalence and abundance of common and rare genera throughout follow up. No consistent associations were identified between breastmilk microbiota composition and ORV outcomes including seroconversion, vaccine shedding after dose 1, and postvaccination rotavirus-specific immunoglobulin A level. CONCLUSIONS Our findings suggest that breastmilk microbiota composition may not be a key factor in shaping trends in ORV response within or between countries.
Collapse
Affiliation(s)
- Jonathan Mandolo
- Virology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Department of Biomedical Sciences, School of Life Sciences and Allied Health Professions, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Edward P K Parker
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Christina Bronowski
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | | | - Alistair C Darby
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Nigel A Cunliffe
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- National Institute for Health and Care Research Health Protection Research Unit in Gastrointestinal Infections, University of Liverpool, Liverpool, United Kingdom
- National Institute for Health and Care Research Global Health Research Group on Gastrointestinal Infections, University of Liverpool, Liverpool, United Kingdom
| | - Gagandeep Kang
- Wellcome Trust Research Laboratory, Division of Gastrointestinal Sciences, Christian Medical College, Vellore, Tamil Nadu, India
| | - Miren Iturriza-Gómara
- Centre for Vaccine Innovation and Access, Program for Appropriate Technology in Health (PATH), Geneva, Switzerland
| | - Arox W Kamng’ona
- Virology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
- Department of Biomedical Sciences, School of Life Sciences and Allied Health Professions, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Khuzwayo C Jere
- Virology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- National Institute for Health and Care Research Health Protection Research Unit in Gastrointestinal Infections, University of Liverpool, Liverpool, United Kingdom
- Department of Medical Laboratory Sciences, School of Life Sciences and Allied Health Professions, Kamuzu University of Health Sciences, Blantyre, Malawi
| |
Collapse
|
8
|
Strobel KM, Juul SE, Hendrixson DT. Maternal Nutritional Status and the Microbiome across the Pregnancy and the Post-Partum Period. Microorganisms 2023; 11:1569. [PMID: 37375071 DOI: 10.3390/microorganisms11061569] [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: 05/15/2023] [Revised: 06/03/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Appropriate nutrition during pregnancy and the post-partum period is vital for both the mothers and their offspring. Both under- and over-nourished status may have important microbial implications on the maternal and infant gut microbiomes. Alterations in the microbiome can have implications for a person's risk of obesity and metabolic diseases. In this review, we examine alterations in the maternal gut, vaginal, placental, and milk microbiomes in the context of pre-pregnancy BMI, gestational weight gain, body composition, gestational diabetes, and maternal diet. We also investigate how the infant gut microbiome may be altered by these different parameters. Many of the microbial changes seen in under- and over-nourished states in birthing parents may result in long-term implications for the health of offspring. Differences in diet appear to be a major driver of the maternal and subsequently milk and offspring microbiomes. Further prospective longitudinal cohort studies are needed to examine nutrition and the microbiome to better understand its implications. Additionally, trials involving dietary interventions in child-bearing age adults should be explored to improve the mother and child's risks for metabolic diseases.
Collapse
Affiliation(s)
- Katie M Strobel
- Department of Pediatrics, University of Washington School of Medicine, 1959 NE Pacific St., Seattle, WA 98195, USA
| | - Sandra E Juul
- Department of Pediatrics, University of Washington School of Medicine, 1959 NE Pacific St., Seattle, WA 98195, USA
| | - David Taylor Hendrixson
- Department of Pediatrics, University of Washington School of Medicine, 1959 NE Pacific St., Seattle, WA 98195, USA
| |
Collapse
|
9
|
Qi C, Tu H, Zhao Y, Zhou J, Chen J, Hu H, Yu R, Sun J. Breast Milk-Derived Limosilactobacillus reuteri Prevents Atopic Dermatitis in Mice via Activating Retinol Absorption and Metabolism in Peyer's Patches. Mol Nutr Food Res 2023; 67:e2200444. [PMID: 36480309 DOI: 10.1002/mnfr.202200444] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 10/24/2022] [Indexed: 12/13/2022]
Abstract
SCOPE Supplementing Limosilactobacillus reuteri Fn041, a breast milk-derived probiotic from agricultural and pastoral areas, to maternal mice during late pregnancy and lactation prevents atopic dermatitis (AD) in offspring. This study aims to elucidate the molecular mechanism of Fn041-mediated immune regulation. METHODS AND RESULTS Fn041 is administered prenatal and postnatal to maternal mice, and to offspring after weaning. The ears are administered with calcipotriol to induce AD. Fn041 treatment significantly alleviates ear inflammation, and reduces mast cell infiltration. Fn041 treatment upregulates and downregulates intestinal ZO-1 and Claudin-2 mRNA expression, respectively. Transcriptome analysis of Peyer's patches reveals that pathways related to DNA damage repair are activated in AD mice, which is inhibited by Fn041 treatment. Fn041 activates pathways related to retinol absorption and metabolism. Untargeted metabolomic analysis reveals that Fn041 treatment increases plasma retinol and kynurenine. Fn041 treatment does not significantly alter the overall cecal microbiota profile, only increases the relative abundances of Ligilactobacillus apodemi, Ligilactobacillus murinus, Akkermansia muciniphila, and Bacteroides thetaiotaomicron. CONCLUSIONS Fn041 induces anti-AD immune responses directly by promoting the absorption and metabolism of retinol in Peyer's patches, and plays an indirect role by strengthening the mucosal barrier and increasing the abundance of specific anti-AD bacteria in the cecum.
Collapse
Affiliation(s)
- Ce Qi
- Institute of Nutrition and Health, Qingdao University, Qingdao, 266071, China
| | - Huayu Tu
- Institute of Nutrition and Health, Qingdao University, Qingdao, 266071, China
| | - Yuning Zhao
- Institute of Nutrition and Health, Qingdao University, Qingdao, 266071, China
| | - Jingbo Zhou
- Institute of Nutrition and Health, Qingdao University, Qingdao, 266071, China
| | - Jie Chen
- Department of Pediatric Cardiology Nephrology and Rheumatism, The Affiliated Hospital of Qingdao University Medical College, Qingdao, 266003, China
| | - Haiting Hu
- Department of Neonatology, The Affiliated Changzhou Maternity and Child Health Care Hospital of Nanjing Medical University, Changzhou, 213004, China
| | - Renqiang Yu
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, 214022, China
| | - Jin Sun
- Institute of Nutrition and Health, Qingdao University, Qingdao, 266071, China
| |
Collapse
|
10
|
Tian M, Li Q, Zheng T, Yang S, Chen F, Guan W, Zhang S. Maternal microbe-specific modulation of the offspring microbiome and development during pregnancy and lactation. Gut Microbes 2023; 15:2206505. [PMID: 37184203 DOI: 10.1080/19490976.2023.2206505] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/16/2023] Open
Abstract
The maternal microbiome is essential for the healthy growth and development of offspring and has long-term effects later in life. Recent advances indicate that the maternal microbiome begins to regulate fetal health and development during pregnancy. Furthermore, the maternal microbiome continues to affect early microbial colonization via birth and breastfeeding. Compelling evidence indicates that the maternal microbiome is involved in the regulation of immune and brain development and affects the risk of related diseases. Modulating offspring development by maternal diet and probiotic intervention during pregnancy and breastfeeding could be a promising therapy in the future. In this review, we summarize and discuss the current understanding of maternal microbiota development, perinatal microbial metabolite transfer, mother-to-infant microbial transmission during/after birth and its association with immune and brain development as well as corresponding diseases.
Collapse
Affiliation(s)
- Min Tian
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qihui Li
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Tenghui Zheng
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Siwang Yang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Fang Chen
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
| | - Wutai Guan
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
| | - Shihai Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
11
|
Sindi AS, Cheema AS, Trevenen ML, Geddes DT, Payne MS, Stinson LF. Characterisation of human milk bacterial DNA profiles in a small cohort of Australian women in relation to infant and maternal factors. PLoS One 2023; 18:e0280960. [PMID: 36696407 PMCID: PMC9876237 DOI: 10.1371/journal.pone.0280960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 01/11/2023] [Indexed: 01/26/2023] Open
Abstract
Human milk is composed of complex microbial and non-microbial components that shape the infant gut microbiome. Although several maternal and infant factors have been associated with human milk microbiota, no study has investigated this in an Australian population. Therefore, we aimed to investigate associations between human milk bacterial composition of Australian women and maternal factors (body mass index (BMI), mode of delivery, breast pump use, allergy, parity) and infant factors (sex, mode of feeding, pacifier use, and introduction of solids). Full-length 16S rRNA gene sequencing was used to characterise milk bacterial DNA profiles. Milk from mothers with a normal BMI had a higher relative abundance of Streptococcus australis than that of underweight mothers, while milk from overweight mothers had a higher relative abundance of Streptococcus salivarius compared with underweight and obese mothers. Mothers who delivered vaginally had a higher relative abundance of Streptococcus mitis in their milk compared to those who delivered via emergency caesarean section. Milk of mothers who used a breast pump had a higher relative abundance of Staphylococcus epidermidis and Streptococcus parasanguinis. Milk of mothers whose infants used a pacifier had a higher relative abundance of S. australis and Streptococcus gwangjuense. Maternal BMI, mode of delivery, breast pump use, and infant pacifier use are associated with the bacterial composition of human milk in an Australian cohort. The data from this pilot study suggests that both mother and infant can contribute to the human milk microbiome.
Collapse
Affiliation(s)
- Azhar S Sindi
- Division of Obstetrics and Gynaecology, School of Medicine, The University of Western Australia, Perth, Western Australia, Australia.,College of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Ali S Cheema
- School of Molecular Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Michelle L Trevenen
- Centre for Applied Statistics, The University of Western Australia, Perth, Western Australia, Australia
| | - Donna T Geddes
- School of Molecular Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Matthew S Payne
- Division of Obstetrics and Gynaecology, School of Medicine, The University of Western Australia, Perth, Western Australia, Australia.,Women and Infants Research Foundation, Perth, Western Australia, Australia
| | - Lisa F Stinson
- School of Molecular Sciences, The University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
12
|
Xu Y, Yang Y, Li B, Xie Y, Shi Y, Le G. Dietary methionine restriction improves gut microbiota composition and prevents cognitive impairment in D-galactose-induced aging mice. Food Funct 2022; 13:12896-12914. [PMID: 36444912 DOI: 10.1039/d2fo03366f] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Dietary methionine restriction (MR) has been shown to delay aging and ameliorate age-related cognitive impairments. We hypothesized that changes in the gut microbiota may mediate these effects. To test this hypothesis, ICR mice subcutaneously injected with 150 mg kg-1 day-1D-galactose were fed a normal (0.86% methionine) or an MR (0.17% methionine) diet for 2 months. Multiple behavioral experiments were performed, and the gut microbiota composition, metabolite profiles related to short-chain fatty acids (SCFAs) in the feces, and indicators related to the redox and inflammatory states in the hippocampus were further analyzed. Our results indicated that MR alleviated cognitive impairment (including non-spatial memory deficits, working memory deficits, and hippocampus-dependent spatial memory deficits) and anxiety-like behavior in D-Gal-induced aging mice. Furthermore, MR increased the abundance of putative SCFA-producing bacteria such as Lachnospiraceae, Turicibacter, Roseburia, Ruminococcaceae_UCG-014, Intestinimonas, Rikenellaceae, Tyzzerella, and H2S-producing bacteria such as Desulfovibrio in feces. Moreover, MR reversed and normalized the levels of intestinal SCFAs (acetate, propionate, and butyrate) and important intermediate metabolites of the SCFAs (pyruvate, lactate, malate, fumarate, and succinate), abolished aging-induced oxidative stress and inflammatory responses, increased the levels of H2S in the plasma and hippocampus, and selectively modulated the expression of multiple learning- and memory-related genes in the hippocampus. These findings suggest that MR improved the gut microbiota composition and SCFA production and alleviated oxidative stress and inflammatory responses in the hippocampus, which might prevent cognitive impairment in D-galactose-induced aging mice.
Collapse
Affiliation(s)
- Yuncong Xu
- National Engineering Laboratory/Key Laboratory of Henan Province, College of Food Science and Engineering, Henan University of Technology, Zhengzhou, Henan, 450001, China. .,State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Yuhui Yang
- National Engineering Laboratory/Key Laboratory of Henan Province, College of Food Science and Engineering, Henan University of Technology, Zhengzhou, Henan, 450001, China.
| | - Bowen Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Yanli Xie
- National Engineering Laboratory/Key Laboratory of Henan Province, College of Food Science and Engineering, Henan University of Technology, Zhengzhou, Henan, 450001, China.
| | - Yonghui Shi
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Guowei Le
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| |
Collapse
|
13
|
Edwards CA, Van Loo-Bouwman CA, Van Diepen JA, Schoemaker MH, Ozanne SE, Venema K, Stanton C, Marinello V, Rueda R, Flourakis M, Gil A, Van der Beek EM. A systematic review of breast milk microbiota composition and the evidence for transfer to and colonisation of the infant gut. Benef Microbes 2022; 13:365-382. [PMID: 36377578 DOI: 10.3920/bm2021.0098] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
Abstract
The intestinal microbiota plays a major role in infant health and development. However, the role of the breastmilk microbiota in infant gut colonisation remains unclear. A systematic review was performed to evaluate the composition of the breastmilk microbiota and evidence for transfer to/colonisation of the infant gut. Searches were performed using PUBMED, OVID, LILACS and PROQUEST from inception until 18th March 2020 with a PUBMED update to December 2021. 88 full texts were evaluated before final critique based on study power, sample contamination avoidance, storage, purification process, DNA extraction/analysis, and consideration of maternal health and other potential confounders. Risk of skin contamination was reduced mainly by breast cleaning and rejecting the first milk drops. Sample storage, DNA extraction and bioinformatics varied. Several studies stored samples under conditions that may selectively impact bacterial DNA preservation, others used preculture reducing reliability. Only 15 studies, with acceptable sample size, handling, extraction, and bacterial analysis, considered transfer of bacteria to the infant. Three reported bacterial transfer from infant to breastmilk. Despite consistent evidence for the breastmilk microbiota, and recent studies using improved methods to investigate factors affecting its composition, few studies adequately considered transfer to the infant gut providing very little evidence for effective impact on gut colonisation.
Collapse
Affiliation(s)
- C A Edwards
- Human Nutrition, School of Medicine, Dentistry, and Nursing, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow Royal Infirmary, 84 Castle St, Glasgow G4 0SF, United Kingdom
| | - C A Van Loo-Bouwman
- Yili Innovation Center Europe, Bronland 12 E-1, 6708 WH Wageningen, the Netherlands
| | - J A Van Diepen
- Medical and Scientific Affairs, Reckitt|Mead Johnson Nutrition Institute, Middenkampweg 2, 6545 CJ Nijmegen, the Netherlands
| | - M H Schoemaker
- Medical and Scientific Affairs, Reckitt|Mead Johnson Nutrition Institute, Middenkampweg 2, 6545 CJ Nijmegen, the Netherlands
| | - S E Ozanne
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, P.O. Box 289, Cambridge CB2 0QQ, United Kingdom
| | - K Venema
- Department of Human Biology, Centre for Healthy Eating & Food Innovation, Maastricht University - Campus Venlo, P.O. Box 8, 5900 AA Venlo, the Netherlands
| | - C Stanton
- Teagasc Moorepark Food Research Centre, and APC Microbiome Ireland, Cork, Ireland
| | - V Marinello
- Human Nutrition, School of Medicine, Dentistry, and Nursing, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow Royal Infirmary, 84 Castle St, Glasgow G4 0SF, United Kingdom
| | - R Rueda
- R&D Department, Abbott Nutrition, Cam. de Purchil, 68, 18004 Granada, Spain
| | - M Flourakis
- ILSI Europe a.i.s.b.l., E. Mounierlaan 83, 1200 Brussels, Belgium; correspondence has been taken over by C.-Y. Chang of ILSI Europe
| | - A Gil
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, Institute of Nutrition and Food Technology 'José Mataix,' Biomedical Research Centre, University of Granada, and Instituto de Investigación Biosanitaria ibs Granada, Avda. del Conocimiento s/n, 18100, Armilla, Grenada, Spain
- CIBEROBN (Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - E M Van der Beek
- Department of Pediatrics, University Medical Centre Groningen, University of Groningen, Postbus 30.001, 9700 RB Groningen, the Netherlands
- Danone Nutricia Research, Uppsalalaan 12, 3584 CT Utrecht, the Netherlands
| |
Collapse
|
14
|
Liu B, Zhao J, Liu Y, Qiao W, Jiang T, Chen L. Diversity and temporal dynamics of breast milk microbiome and its influencing factors in Chinese women during the first 6 months postpartum. Front Microbiol 2022; 13:1016759. [DOI: 10.3389/fmicb.2022.1016759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 10/19/2022] [Indexed: 11/13/2022] Open
Abstract
Human breast milk (HBM) plays an important role in providing nutrients, beneficial microorganisms and bioactive components for infants, helping maturation of their immune system and gastrointestinal development. Here, we present a study aiming to investigate the diversity and temporal dynamics of the milk microbiome across the first 6 month postpartum in Chinese healthy breastfeeding women, and to investigate to what extent other variables (e.g., sampling location, infant sex, and mode of delivery) might also be related to variations in the human milk microbiome, and the association with maternal diet and nutrients. Fifty-three healthy pregnant women from four cities were recruited from a China Maternal and Infant Health Cohort Study and breast milk samples were collected and analyzed using 16S rRNA metagenomic sequencing. We illustrated the diversity and temporal dynamics during lactation (Adonis p-value = 3e–04). Firmicutes and Proteobacteria were the most abundant phyla, and Streptococcus, Staphylococcus, Serratia, and Corynebacterium were the core genera. Partitioning around medoids clustering identified two major internal clusters of breast milk microbiota. Cluster 1 was dominated by Acinetobacter and Pseudomonas, while Cluster 2 was dominated by Streptococcus and Staphylococcus. Among other environmental variables, sampling location showed significant influence on breast milk microbiome (Adonis p-value = 4e–04), while infant sex (Adonis p-value = 0.33) and mode of delivery (Adonis p-value = 0.19) were less related to variations in the human milk microbiome. Maternal diet such as tuber was significantly correlated with the relative abundance of Neisseria (rho = 0.34, adjusted p-value = 0.01) and Cutibacterium (rho = −0.35, adjusted p-value = 0.01), and nutrients such as carbohydrates were significantly correlated with the relative abundance of Aquabacterium (rho = −0.39, adjusted p-value = 0.0027), and vitamin B12 was significantly correlated with the relative abundance of Coprococcus (rho = 0.40, adjusted p-value = 0.0018), etc. These results illustrated the dynamic changes of composition and diversity during the lactation phases of the Chinese breast milk microbiome and addressed the importance of geographic location on milk microbiota, and associations with maternal diet consumption, which have potential benefits on the establishment and future health of breastfeeding infants.
Collapse
|
15
|
Maternal weight status and the composition of the human milk microbiome: A scoping review. PLoS One 2022; 17:e0274950. [PMID: 36191014 PMCID: PMC9529148 DOI: 10.1371/journal.pone.0274950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 09/07/2022] [Indexed: 11/19/2022] Open
Abstract
The human milk microbiome is thought to partly contribute to the assembly of the infant gut microbiome, a microbial community with important implications for infant health and development. While obesity has well-established links with the adult gut microbiome, less is known about how it affects the human milk microbiome. In this scoping review, we synthesize the current literature on the microbial composition of human milk by maternal weight status, defined broadly as BMI (prepregnancy and postpartum) and gestational weight gain (GWG). This study followed the a priori protocol published in Prospero (registration #: CRD42020165633). We searched the following databases for studies reporting maternal weight status and a characterization of milk microbiota through culture-dependent and culture-independent methods: MEDLINE, Embase, Web of Science, CINAHL, and Scopus. After screening 6,365 studies, we found 20 longitudinal and cross-sectional studies investigating associations between maternal weight status and the composition of the milk microbiome. While some studies reported no associations, many others reported that women with a pre-pregnancy or postpartum BMI characterized as overweight or obese, or with excessive GWG, had higher abundances of the genus Staphylococcus, lower Bifidobacterium abundance, and lower alpha diversity (within-sample diversity). This review suggests that maternal weight status is minorly associated with the composition of the milk microbiome in various ways. We offer potential explanations for these findings, as well as suggestions for future research.
Collapse
|
16
|
Du Y, Qiu Q, Cheng J, Huang Z, Xie R, Wang L, Wang X, Han Z, Jin G. Comparative study on the microbiota of colostrum and nipple skin from lactating mothers separated from their newborn at birth in China. Front Microbiol 2022; 13:932495. [PMID: 36262322 PMCID: PMC9574262 DOI: 10.3389/fmicb.2022.932495] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 09/02/2022] [Indexed: 11/13/2022] Open
Abstract
Increasing studies have found breast milk (BM) contains its own microbiota. However, the route through which microbes enter the BM is still unclear. In order to verify the entero-mammary pathway of BM, we designed a rigorous study that prevented oral bacteria from contaminating the breast and nipple skin (NS) during baby nursing. Thirty-one healthy, postpartum mothers living in southern China who were immediately separated from their newborn after delivery were enrolled in this study. Using an aseptic protocol for sampling, sterile water was used to wash the NS and was then collected. Then the first drop of BM was discarded and colostrum was collected manually. Amplicon sequencing was performed targeting the V3–V4 region of the bacterial 16S rRNA gene, and the differences between the microbiota of the colostrum and NS were analyzed. Additionally, the effects of environmental factors, such as the delivery mode and intrapartum antibiotic exposure, on the diversity of the colostrum microbiota were also analyzed. We found significant differences in the α diversity and richness between the BM and NS as evidenced by richness, Chao1, and Simpson indices. There were 170 operational taxonomic units (OTUs) shared by colostrum and NS, while 111 and 87 OTUs were unique, respectively, as well as a clear distinction in OTUs was observed by unifrac binary analysis between them. Linear discriminant analysis effect size analysis found that anaerobes, such as Bifidobacterium and Pantoea at the genus level and enterobacteria including Enterobacteriaceae at the family level, were predominant in the colostrum, while the predominant bacteria on the NS were Bacteroides, Staphylococcus, and Parabacteroides at the genus level. BM is colonized by bacteria prior to baby suckling, and the diversity of the colostrum microbiota differs from that of the NS. The predominant microbiota taxa in BM indicated that they were likely to be transferred to the breast through the intestinal tract. Our study provides direct evidence for the revolutionary active migration hypothesis. Additionally, factors like intrapartum antibiotic exposure did not significantly affect the diversity of the microbiota in the BM. Therefore, it is suggested that mothers continue to provide BM for their newborns during separation.
Collapse
Affiliation(s)
- Yanli Du
- School of Medical Technology and Nursing, Shenzhen Polytechnic, Shenzhen, China
| | - Qing Qiu
- Department of Women Health Care, Shenzhen Luohu Maternity and Child Healthcare Hospital, Shenzhen, China
| | - Jing Cheng
- Department of Obstetrics, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Zhili Huang
- School of Medical Technology and Nursing, Shenzhen Polytechnic, Shenzhen, China
| | - Ruixia Xie
- School of Medical Technology and Nursing, Shenzhen Polytechnic, Shenzhen, China
| | - Lu Wang
- Delivery Center, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, China
| | - Xiangyu Wang
- Shenzhen Second People’s Hospital, Department of Gastroenterology, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
- Xiangyu Wang,
| | - Zongli Han
- Department of Neurosurgery, Peking University Shenzhen Hospital, Shenzhen, China
- *Correspondence: Zongli Han,
| | - Gang Jin
- School of Medical Technology and Nursing, Shenzhen Polytechnic, Shenzhen, China
- Gang Jin,
| |
Collapse
|
17
|
Zhou J, Xu G, Li X, Tu H, Li H, Chang H, Chen J, Yu R, Qi C, Sun J. Limosilactobacillus reuteri FN041 prevents atopic dermatitis in pup mice by remodeling the ileal microbiota and regulating gene expression in Peyer’s patches after vertical transmission. Front Nutr 2022; 9:987400. [PMID: 36245510 PMCID: PMC9554658 DOI: 10.3389/fnut.2022.987400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/12/2022] [Indexed: 12/04/2022] Open
Abstract
Objectives Limosilactobacillus reuteri FN041 is a potential probiotic bacterium isolated from breast milk in traditional farming and pastoral areas of China. The purpose of this study was to investigate the optimal intervention mode and potential mechanism of FN041 to prevent atopic dermatitis (AD) in mice. Methods In intervention mode I, FN041 was supplemented to dams during the late trimester and lactation and pups after weaning; in intervention mode II, FN041 was supplemented after pups were weaned. AD was induced in pups with MC903 plus ovalbumin on the ear after weaning. Results The effect of intervention mode I in preventing AD was significantly better than that of intervention mode II. Compared with the model group, the inflammatory response of the pup’s ears, the proportion of spleen regulatory T cells and the plasma IgE were significantly decreased in mice in intervention mode I. Furthermore, the intestinal mucosal barrier was enhanced, and the Shannon index of the ileal microbiota was significantly increased. The microbiota structure deviated from the AD controls and shifted toward the healthy controls according to the PCoA of unweighted UniFrac. The relative abundances of Limosilactobacillus, Faecalibacterium, Bifidobacterium, and Akkermansia in the ileum were significantly increased compared to the AD group. Based on RNA-seq analysis of pups’ Peyer’s patches (PPs), FN041 inhibits autoimmune pathways such as asthma and systemic lupus erythematosus and activates retinol metabolism and PPAR signaling pathways to reduce inflammatory responses. Intervention mode II also significantly reduced AD severity score, but the reduction was approximately 67% of that of intervention mode I. This may be related to its ineffective remodeling of the ileal microbiota. Conclusion Prenatal and postnatal administration of FN041 is an effective way to prevent AD in offspring, and its mechanism is related to remodeling of ileal microbiota and PPs immune response.
Collapse
Affiliation(s)
- Jingbo Zhou
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Gaoshun Xu
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Xinyue Li
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Huayu Tu
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Haoyu Li
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Hong Chang
- Department of Pediatric Cardiology Nephrology and Rheumatism, The Affiliated Hospital of Qingdao University Medical College, Qingdao, China
| | - Jie Chen
- Department of Pediatric Cardiology Nephrology and Rheumatism, The Affiliated Hospital of Qingdao University Medical College, Qingdao, China
| | - Renqiang Yu
- Department of Neonatology, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, China
- Renqiang Yu,
| | - Ce Qi
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
- Ce Qi,
| | - Jin Sun
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
- *Correspondence: Jin Sun,
| |
Collapse
|
18
|
Stinson LF, Ma J, Sindi AS, Geddes DT. Methodological approaches for studying the human milk microbiome. Nutr Rev 2022; 81:705-715. [PMID: 36130405 DOI: 10.1093/nutrit/nuac082] [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: 11/13/2022] Open
Abstract
Human milk contains a low-biomass, low-diversity microbiome, consisting largely of bacteria. This community is of great research interest in the context of infant health and maternal and mammary health. However, this sample type presents many unique methodological challenges. In particular, there are numerous technical considerations relating to sample collection and storage, DNA extraction and sequencing, viability, and contamination. Failure to properly address these challenges may lead to distortion of bacterial DNA profiles generated from human milk samples, ultimately leading to spurious conclusions. Here, these technical challenges are discussed, and various methodological approaches used to address them are analyzed. Data were collected from studies in which a breadth of methodological approaches were used, and recommendations for robust and reproducible analysis of the human milk microbiome are proposed. Such methods will ensure high-quality data are produced in this field, ultimately supporting better research outcomes for mothers and infants.
Collapse
Affiliation(s)
- Lisa F Stinson
- School of Molecular Sciences, The University of Western Australia, Perth, Australia
| | - Jie Ma
- School of Molecular Sciences, The University of Western Australia, Perth, Australia
| | - Azhar S Sindi
- Division of Obstetrics and Gynaecology, School of Medicine, The University of Western Australia, Perth, Australia.,is with the 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, Australia
| |
Collapse
|
19
|
Lopez Leyva L, Gonzalez E, Solomons NW, Koski KG. Human milk microbiome is shaped by breastfeeding practices. Front Microbiol 2022; 13:885588. [PMID: 36160202 PMCID: PMC9493375 DOI: 10.3389/fmicb.2022.885588] [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: 02/28/2022] [Accepted: 08/08/2022] [Indexed: 11/21/2022] Open
Abstract
There is evidence that breastfeeding practices may impact the milk microbiota diversity and differential abundance at the genera level; however, the possibility that distinct feeding practices, such as exclusive (EBF) and non-exclusive breastfeeding (non-EBF), might alter the milk microbiome at the species level has not been explored. This cross-sectional study analyzed the milk microbiome of 64 Mam-Mayan indigenous mothers from San Juan Ostuncalco in Guatemala. Two breastfeeding practices [exclusive (EBF) vs non-exclusive (non-EBF)] were analyzed at two stages of lactation [early (5–46 days post-partum) vs late (109–184 days post-partum)]. EBF was defined as offering only human milk and non-EBF was defined as feeding the infant herbal teas (agüitas) and/or complementary foods while continuing to breastfeed. Results identified four clusters with distinct microbial communities that segregated bacterial species by both breastfeeding practices and stage of lactation. Comparison among these clusters identified several notable patterns. First, during EBF, the microbiome differed by stage of lactation where there was a shift in differential abundance from Actinobacteria and Firmicutes in early to Bacteroidetes and Proteobacteria species in late lactation. Second, a similar comparison between non-EBF mothers by stage of lactation also identified a higher differential abundance of Actinobacteria and Firmicutes species in early lactation, but only Proteobacteria and not Bacteroidetes in late lactation, indicating a further shift in the milk microbial ecosystem with fewer oral bacteria present in late lactation. Third, comparisons between EBF and non-EBF mothers at both early and late lactation showed that mothers who exclusively breastfed had more differentially abundant species in early (11 vs 1) and late (13 vs 2) lactation. Fourth, EBF at early and late lactation had more commensal and lactic acid bacteria, including Lactobacillus gasseri, Granulicatella elegans, Streptococcus mitis, and Streptococcus parasanguinis, compared to those who did not exclusively breastfeed. Collectively, these results show that EBF has more differentially abundant bacteria, including commensal and lactic acid bacteria, and that the addition of agüitas (herbal teas) and/or complementary foods modify the milk microbiome composition by reducing the oral bacteria and introducing more environmentally sourced bacteria to the ecosystem.
Collapse
Affiliation(s)
| | - Emmanuel Gonzalez
- Canadian Centre for Computational Genomics (C3G), Department of Human Genetics, McGill University, Montréal, QC, Canada
- Microbiome Research Platform, McGill Interdisciplinary Initiative in Infection and Immunity (MI4), Genome Centre, McGill University, Montreal, QC, Canada
| | - Noel W. Solomons
- Center for Studies of Sensory Impairment, Aging and Metabolism (CeSSIAM), Guatemala City, Guatemala
| | - Kristine G. Koski
- School of Human Nutrition, McGill University, Montreal, QC, Canada
- *Correspondence: Kristine G. Koski,
| |
Collapse
|
20
|
Ding M, Chen H, Yu R, Ross RP, Stanton C, Zhang H, Yang B, Chen W. Shared and Non-Shared sIgA-Coated and -Uncoated Bacteria in Intestine of Mother–Infant Pairs. Int J Mol Sci 2022; 23:ijms23179873. [PMID: 36077271 PMCID: PMC9456154 DOI: 10.3390/ijms23179873] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/19/2022] [Accepted: 08/26/2022] [Indexed: 11/23/2022] Open
Abstract
The infant gut microbiota is critical for promoting and maintaining early-life health. The study aimed to analyze the composition of sIgA-coated and sIgA-uncoated bacterial communities at genus level and lactobacilli and bifidobacterial communities at species level in human breast milk (HBM) and infant and maternal feces. Eleven pregnant women were recruited successfully. HBM; infant feces during colostrum, transition, and mature stages; and maternal feces within the mature stage were collected. sIgA-coated and sIgA-uncoated bacteria were separated with magnetic-activated cell sorting. Then, 16S rRNA sequencing, bifidobacterial groEL gene sequencing, and lactobacilli groEL gene sequencing were performed to analyze the bacterial community. PCoA revealed that the compositions of sIgA-coated and sIgA-uncoated bacteria were different among HBM and infant and maternal feces. Higher relative abundance of sIgA-uncoated Bifidobacterium was found in the three lactation stages in infant feces compared to the corresponding HBM, and a higher relative abundance of sIgA-uncoated Faecalibacterium was found in maternal feces compared to HBM and infant feces. For bifidobacterial community, sIgA-coated and sIgA-uncoated B. longum subsp. infantis and B. pseudocatenulatum was dominant in infant feces and maternal feces, respectively. The relative abundance of sIgA-uncoated B. longum subsp. infantis was significantly higher in infant feces compared to that in maternal feces. For the Lactobacillus community, L. paragasseri and L. mucosae were dominant in infant and maternal feces, respectively. HBM and infant and maternal feces showed distinct diversity and composition of both sIgA-coated and sIgA-uncoated bacteria at genus level. Infant and maternal feces showed similar composition of Bifidobacterium at species level. The same Bifidobacterium species could be detected both in sIgA-coated and -uncoated form. This article provided deeper understanding on the microbiota profile in HBM and infant and maternal feces.
Collapse
Affiliation(s)
- Mengfan Ding
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Haiqin Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Renqiang Yu
- Department of Neonatology, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi 214122, China
- Correspondence: (R.Y.); (B.Y.)
| | - Reynolds Paul Ross
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi 214122, China
- APC Microbiome Ireland, University College Cork, T12 K8AF Cork, Ireland
| | - Catherine Stanton
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi 214122, China
- APC Microbiome Ireland, University College Cork, T12 K8AF Cork, Ireland
- Teagasc Food Research Centre, Moorepark, Fermoy, P61 C996 Co Cork, Ireland
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
- Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi 214122, China
| | - Bo Yang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi 214122, China
- Correspondence: (R.Y.); (B.Y.)
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi 214122, China
| |
Collapse
|
21
|
Ding M, Zheng Y, Liu F, Tian F, Ross RP, Stanton C, Yu R, Zhao J, Zhang H, Yang B, Chen W. Lactation time influences the composition of Bifidobacterium and Lactobacillus at species level in human breast milk. Benef Microbes 2022; 13:319-330. [PMID: 35979712 DOI: 10.3920/bm2021.0119] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Human breast milk is a source of microorganisms for infants that play an important role in building infant gut health and immunity. The bacterial composition in human breast milk is influenced by lactation time. This study aimed to investigate the influence of lactation time on bacteria in breast milk at the genus level and the species levels of Bifidobacterium and Lactobacillus on days 2-4, 8, 14, and 30. Eighteen individuals were recruited and 60 milk samples were collected. The 16S rRNA gene, and the bifidobacterial groEL and lactobacilli groEL genes were used for amplicon sequencing. The results revealed that the alpha diversities of colostrum and transition 1 (day 8) milk were lower than that of transition 2 (day 14) and mature milk. PCoA analysis showed that bacterial composition in colostrum and transition 1 milk differed from transition 2 and mature milk. A lower relative abundance of Blautia was found in colostrum and transition 1 milk compared with mature milk and lower abundances of Ruminococcus, Dorea, and Escherichia-Shigella were found in transition 1 compared with mature milk. Bifidobacterium ruminantium, Limosilactobacillus mucosae, and Ligilactobacillus ruminis were the predominant species across all four lactation stages, while Bifidobacterium bifidum was lower in transition 1, and Bifidobacterium pseudocatenulatum and Bifidobacterium pseudolongum were higher in transition 1 milk. This study indicated that the bacterial composition in colostrum was more similar to that of transition 1 milk, whereas the bacterial community in transition 2 milk was similar to that of mature milk which suggests that bacterial composition in human breast milk shows stage-specific signatures even within a short period at both genus level and Bifidobacterium and Lactobacillus species levels, providing insights into probiotic supplementation for the nursing mother.
Collapse
Affiliation(s)
- M Ding
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China P.R.,School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122 Jiangsu, China P.R
| | - Y Zheng
- H&H Global Research and Technology Center, Guangzhou, China P.R
| | - F Liu
- H&H Global Research and Technology Center, Guangzhou, China P.R
| | - F Tian
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China P.R.,School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122 Jiangsu, China P.R
| | - R P Ross
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi, Jiangsu, China P.R.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - C Stanton
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi, Jiangsu, China P.R.,APC Microbiome Ireland, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Fermoy, Cork P61 C996, Ireland
| | - R Yu
- Department of Neonatology, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University,48 Huaishu Alley, Liangxi District, Wuxi, 214002, China P.R
| | - J Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China P.R.,School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122 Jiangsu, China P.R
| | - H Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China P.R.,School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122 Jiangsu, China P.R.,National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China P.R.,Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, China P.R
| | - B Yang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China P.R.,School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122 Jiangsu, China P.R.,H&H Global Research and Technology Center, Guangzhou, China P.R
| | - W Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China P.R.,School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122 Jiangsu, China P.R.,National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China P.R
| |
Collapse
|
22
|
Gómez-Torres N, Sánchez-García L, Castro I, Arroyo R, Cabañas F, González-Sánchez R, López-Azorín M, Moral-Pumarega MT, Escuder-Vieco D, Cabañes-Alonso E, Rodríguez JM, Alba C, Pellicer A. Metataxonomic Analysis of Milk Samples From SARS-CoV-2-Positive and SARS-CoV-2-Negative Women. Front Nutr 2022; 9:853576. [PMID: 35369105 PMCID: PMC8971750 DOI: 10.3389/fnut.2022.853576] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/24/2022] [Indexed: 12/12/2022] Open
Abstract
Objective To assess the impact of SARS-CoV-2 viral infection on the metataxonomic profile and its evolution during the first month of lactation. Methods Milk samples from 37 women with full-term pregnancies and mild SARS-CoV-2 infection and from 63 controls, collected in the first and fifth postpartum weeks, have been analyzed. SARS-CoV-2 RNA was assessed by reverse transcription polymerase chain reaction (RT-PCR) both in cases and controls. After DNA extraction, the V3-V4 hypervariable region of the gene 16S rRNA was amplified and sequenced using the MiSeq system of Illumina. Data were submitted for statistical and bioinformatics analyses after quality control. Results All the 1st week and 5th week postpartum milk samples were negative for SARS-CoV-2 RNA. Alpha diversity showed no differences between milk samples from the study and control group, and this condition was maintained along the observation time. Analysis of the beta-diversity also indicated that the study and control groups did not show distinct bacterial profiles. Staphyloccus and Streptococcus were the most abundant genera and the only ones that were detected in all the milk samples provided. Disease state (symptomatic or asymptomatic infection) did not affect the metataxonomic profile in breast milk. Conclusion These results support that in the non-severe SARS-CoV-2 pregnant woman infection the structure of the bacterial population is preserved and does not negatively impact on the human milk microbiota.
Collapse
Affiliation(s)
- Natalia Gómez-Torres
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| | - Laura Sánchez-García
- Department of Neonatology, Biomedical Research Foundation-IDIPAZ, La Paz University Hospital, Madrid, Spain
- *Correspondence: Laura Sánchez-García,
| | - Irma Castro
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| | - Rebeca Arroyo
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| | - Fernando Cabañas
- Department of Neonatology, Quirónsalud Madrid University Hospital and Quirónsalud San José Hospital, Biomedical Research Foundation-IDIPAZ, La Paz University Hospital, Madrid, Spain
| | - Raquel González-Sánchez
- Department of Neonatology, Quirónsalud Madrid University Hospital and Quiroónsalud San José Hospital, Madrid, Spain
| | - Manuela López-Azorín
- Department of Neonatology, Quirónsalud Madrid University Hospital and Quiroónsalud San José Hospital, Madrid, Spain
| | | | | | - Esther Cabañes-Alonso
- Department of Neonatology, Regional Human Milk Bank, 12 de Octubre University Hospital, Madrid, Spain
| | - Juan Miguel Rodríguez
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| | - Claudio Alba
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
- Claudio Alba,
| | - Adelina Pellicer
- Department of Neonatology, Biomedical Research Foundation-IDIPAZ, La Paz University Hospital, Madrid, Spain
- *Correspondence: Laura Sánchez-García,
| |
Collapse
|
23
|
Lu M, Sun J, Zhao Y, Zhang H, Li X, Zhou J, Dang H, Zhang J, Huang W, Qi C, Li D. Prevention of High-Fat Diet-Induced Hypercholesterolemia by Lactobacillus reuteri Fn041 Through Promoting Cholesterol and Bile Salt Excretion and Intestinal Mucosal Barrier Functions. Front Nutr 2022; 9:851541. [PMID: 35369106 PMCID: PMC8967143 DOI: 10.3389/fnut.2022.851541] [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: 01/10/2022] [Accepted: 02/17/2022] [Indexed: 12/12/2022] Open
Abstract
Objectives: Lactobacillus reuteri Fn041 (Fn041) is a probiotic isolated from immunoglobulin A coated microbiota in the human breast milk of Gannan in China with a low incidence of hypercholesterolemia. This study aims to explore the role and mechanism of Fn041 in preventing hypercholesterolemia caused by a high-fat diet in mice. Methods C57BL/6N mice were fed a low-fat diet or a high-fat diet and gavage with Fn041 and Lactobacillus rhamnosus GG (LGG) for 8 weeks. Results Both Fn041 and LGG prevented the occurrence of hypercholesterolemia, liver and testicular fat accumulation. In addition, a high-fat diet causes intestinal dysbiosis and mucosal barrier damage, which is associated with hypercholesterolemia. Fn041 prevented the high-fat diet-induced reduction in alpha diversity of intestinal microbiota and intestinal mucosal barrier damage. Fn041 treatment significantly increased fecal total cholesterol and total bile acids. Conclusions Fn041 prevented hypercholesterolemia by enhancing cholesterol excretion and mucosal barrier function.
Collapse
Affiliation(s)
- Mengyao Lu
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Jin Sun
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Yuning Zhao
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Haowen Zhang
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Xinyue Li
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Jingbo Zhou
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Hongyang Dang
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Jidong Zhang
- Department of Cardiology, The Affiliated Hospital of Medical College, Qingdao University, Qingdao, China
| | - Wenjing Huang
- Department of Paediatrics, The Affiliated Hospital of Medical College, Qingdao University, Qingdao, China
| | - Ce Qi
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
- *Correspondence: Ce Qi
| | - Duo Li
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
- Duo Li
| |
Collapse
|
24
|
Yuan L, Zhang X, Luo B, Li X, Tian F, Yan W, Ni Y. Ethnic Specificity of Species and Strain Composition of Lactobacillus Populations From Mother–Infant Pairs, Uncovered by Multilocus Sequence Typing. Front Microbiol 2022; 13:814284. [PMID: 35387090 PMCID: PMC8979337 DOI: 10.3389/fmicb.2022.814284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 01/03/2022] [Indexed: 11/13/2022] Open
Abstract
The maternal gut is thought to be the principal source of potential probiotic bacteria in the infant gut during the lactation stage. It is not clear whether facultative symbiont lactobacilli strictly follow vertical transmission from mother to infant and display the ethnic specificity in terms of species and strain composition in mother–infant cohorts. In the present study, a total of 16 former Lactobacillus species (365 strains) and 11 species (280 strains) were retrieved from 31 healthy mother–infant pairs of two ethnic groups, which have never intermarried, respectively. The result showed that the composition and number of Lactobacillus species between the two ethnic groups varied. Among 106 Lacticaseibacillus paracasei strains isolated, 64 representative strains were classified into 27 sequence types (ST) by means of multilocus sequence typing (MLST), of which 20 STs derived from 33 Uighur strains and 7 STs from 31 Li strains, and no homologous recombination event of genes was detected between strains of different ethnic groups. A go-EBURST analysis revealed that except for a few mother–infant pairs in which more than one STs were detected, L. paracasei isolates from the same mother–infant pair were found to be monophyletic in most cases, confirming vertical transfer of Lactobacillus at the strain level. More notably, L. paracasei isolates from the same ethnic group were more likely than strains from another to be incorporated into a specific phylogenetic clade or clonal complex (CC) with similar metabolic profile of glycan, supporting the hypothesis of ethnic specificity to a large degree. Our study provides evidence for the development of personalized probiotic tailored to very homogenous localized populations from the perspective of maternal and child health.
Collapse
Affiliation(s)
- Lixia Yuan
- School of Food Science and Technology, Shihezi University, Shihezi, China
| | - Xueling Zhang
- School of Food Science and Technology, Shihezi University, Shihezi, China
| | - Baolong Luo
- School of Food Science and Technology, Shihezi University, Shihezi, China
| | - Xu Li
- School of Food Science and Technology, Shihezi University, Shihezi, China
| | - Fengwei Tian
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Wenli Yan
- School of Food Science and Technology, Shihezi University, Shihezi, China
- *Correspondence: Wenli Yan,
| | - Yongqing Ni
- School of Food Science and Technology, Shihezi University, Shihezi, China
- Yongqing Ni,
| |
Collapse
|
25
|
Cheema AS, Trevenen ML, Turlach BA, Furst AJ, Roman AS, Bode L, Gridneva Z, Lai CT, Stinson LF, Payne MS, Geddes DT. Exclusively Breastfed Infant Microbiota Develops over Time and Is Associated with Human Milk Oligosaccharide Intakes. Int J Mol Sci 2022; 23:2804. [PMID: 35269946 PMCID: PMC8910998 DOI: 10.3390/ijms23052804] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/22/2022] [Accepted: 02/28/2022] [Indexed: 12/11/2022] Open
Abstract
Temporal development of maternal and infant microbiomes during early life impacts short- and long-term infant health. This study aimed to characterize bacterial dynamics within maternal faecal, human milk (HM), infant oral, and infant faecal samples during the exclusive breastfeeding period and to document associations between human milk oligosaccharide (HMO) intakes and infant oral and faecal bacterial profiles. Maternal and infant samples (n = 10) were collected at 2−5, 30, 60, 90 and 120 days postpartum and the full-length 16S ribosomal RNA (rRNA) gene was sequenced. Nineteen HMOs were quantitated using high-performance liquid chromatography. Bacterial profiles were unique to each sample type and changed significantly over time, with a large degree of intra- and inter-individual variation in all sample types. Beta diversity was stable over time within infant faecal, maternal faecal and HM samples, however, the infant oral microbiota at day 2−5 significantly differed from all other time points (all p < 0.02). HMO concentrations and intakes significantly differed over time, and HMO intakes showed differential associations with taxa observed in infant oral and faecal samples. The direct clinical relevance of this, however, is unknown. Regardless, future studies should account for intakes of HMOs when modelling the impact of HM on infant growth, as it may have implications for infant microbiota development.
Collapse
Affiliation(s)
- Ali Sadiq Cheema
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (A.S.C.); (Z.G.); (C.T.L.); (L.F.S.)
| | - Michelle Louise Trevenen
- Centre for Applied Statistics, The University of Western Australia, Crawley, WA 6009, Australia; (M.L.T.); (B.A.T.)
| | - Berwin Ashoka Turlach
- Centre for Applied Statistics, The University of Western Australia, Crawley, WA 6009, Australia; (M.L.T.); (B.A.T.)
| | - Annalee June Furst
- Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence, University of California San Diego, La Jolla, CA 92093, USA; (A.J.F.); (A.S.R.); (L.B.)
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| | - Ana Sophia Roman
- Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence, University of California San Diego, La Jolla, CA 92093, USA; (A.J.F.); (A.S.R.); (L.B.)
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| | - Lars Bode
- Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence, University of California San Diego, La Jolla, CA 92093, USA; (A.J.F.); (A.S.R.); (L.B.)
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| | - Zoya Gridneva
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (A.S.C.); (Z.G.); (C.T.L.); (L.F.S.)
| | - Ching Tat Lai
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (A.S.C.); (Z.G.); (C.T.L.); (L.F.S.)
| | - Lisa Faye Stinson
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (A.S.C.); (Z.G.); (C.T.L.); (L.F.S.)
| | - Matthew Scott Payne
- Division of Obstetrics and Gynaecology, School of Medicine, The University of Western Australia, Subiaco, WA 6008, Australia;
- Women and Infants Research Foundation, Subiaco, WA 6008, Australia
| | - Donna Tracy Geddes
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (A.S.C.); (Z.G.); (C.T.L.); (L.F.S.)
| |
Collapse
|
26
|
Yang B, Ding M, Chen Y, Han F, Yang C, Zhao J, Malard P, Stanton C, Ross RP, Zhang H, Chen W. Development of gut microbiota and bifidobacterial communities of neonates in the first 6 weeks and their inheritance from mother. Gut Microbes 2022; 13:1-13. [PMID: 33847206 PMCID: PMC8049200 DOI: 10.1080/19490976.2021.1908100] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [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
Microbiota especially Bifidobacterium play an important role in adjusting and maintaining homeostatic balance within the infant intestine. The aim of this study was to elucidate the relationship between maternal and infant gut microbiota and identify the Bifidobacterium species that may transfer from mother to infant over the first 42 days of the infant's life. Nineteen mother-infant-pair fecal samples were collected and the diversity and composition of the total bacterial and Bifidobacterium communities were analyzed via 16S rDNA and bifidobacterial groEL gene high throughput sequencing. The results revealed that the relative abundance of Bifidobacterium was significantly higher in the infant gut while Parabacteroides, Blautia, Coprococcus, Lachnospira and Faecalibacterium were at lower relative abundance in 7-day and 42-day infant fecal samples compared to the maternal samples. The maternal gut has more B. pseudocatenulatum. In the infant group, B. breve and B. dentium relative abundance increased while B. animalis subsp. lactis decreased from days 7 to 42. Additionally, B. longum subsp. longum isolated from FGZ16 and FGZ35 may have transferred from mother to infant and colonized the infant gut. The results of the current study provide insight toward the infant gut microbiota composition and structure during the first 42 days and may help guide Bifidobacterium supplementation strategies in mothers and infants.
Collapse
Affiliation(s)
- Bo Yang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China,School of Food Science and Technology, Jiangnan University, Wuxi, China,International Joint Research Laboratory for Pharmabiotics & Antibiotic Resistance, Jiangnan University, Wuxi, China
| | - Mengfan Ding
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China,School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Yingqi Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China,School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Fengzhen Han
- Department of Gynaecology and Obsterics, Guangdong Province People’s Hospital, Guangdong Academy of Medical Science, Guangzhou, China
| | - Chunyan Yang
- Department of Gynaecology and Obsterics, Guangdong Province People’s Hospital, Guangdong Academy of Medical Science, Guangzhou, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China,School of Food Science and Technology, Jiangnan University, Wuxi, China,National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| | - Patrice Malard
- Biostime (Guangzhou) Health Products Ltd., Guangzhou, China
| | - Catherine Stanton
- International Joint Research Laboratory for Pharmabiotics & Antibiotic Resistance, Jiangnan University, Wuxi, China,Food Bioscience, Teagasc Food Research Centre, Fermoy, Ireland,CONTACT Catherine Stanton Teagasc Food Research Centre, Fermoy, Ireland
| | - R. Paul Ross
- International Joint Research Laboratory for Pharmabiotics & Antibiotic Resistance, Jiangnan University, Wuxi, China,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China,School of Food Science and Technology, Jiangnan University, Wuxi, China,National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China,Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China,School of Food Science and Technology, Jiangnan University, Wuxi, China,National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China,Wei Chen School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
27
|
Gámez-Valdez JS, García-Mazcorro JF, Montoya-Rincón AH, Rodríguez-Reyes DL, Jiménez-Blanco G, Rodríguez MTA, de Vaca RPC, Alcorta-García MR, Brunck M, Lara-Díaz VJ, Licona-Cassani C. Differential analysis of the bacterial community in colostrum samples from women with gestational diabetes mellitus and obesity. Sci Rep 2021; 11:24373. [PMID: 34934118 PMCID: PMC8692321 DOI: 10.1038/s41598-021-03779-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 12/06/2021] [Indexed: 02/07/2023] Open
Abstract
Gestational Diabetes Mellitus (GDM) and obesity affect the functioning of multiple maternal systems and influence colonization of the newborn gastrointestinal through the breastmilk microbiota (BMM). It is currently unclear how GDM and obesity affect the human BMM composition. Here, we applied 16S-rRNA high-throughput sequencing to human colostrum milk to characterize BMM taxonomic changes in a cohort of 43 individuals classified in six subgroups according to mothers patho-physiological conditions (healthy control (n = 18), GDM (n = 13), or obesity (n = 12)) and newborn gender. Using various diversity indicators, including Shannon/Faith phylogenetic index and UniFrac/robust Aitchison distances, we evidenced that BMM composition was influenced by the infant gender in the obesity subgroup. In addition, the GDM group presented higher microbial diversity compared to the control group. Staphylococcus, Corynebacterium 1, Anaerococcus and Prevotella were overrepresented in colostrum from women with either obesity or GDM, compared to control samples. Finally, Rhodobacteraceae was distinct for GDM and 5 families (Bdellovibrionaceae, Halomonadaceae, Shewanellaceae, Saccharimonadales and Vibrionaceae) were distinct for obesity subgroups with an absolute effect size greater than 1 and a q-value ≤ 0.05. This study represents the first effort to describe the impact of maternal GDM and obesity on BMM.
Collapse
Affiliation(s)
- J S Gámez-Valdez
- Tecnológico de Monterrey, Escuela de Ingeniería y Ciencias, Ave. Eugenio Garza Sada 2501 sur, Monterrey, NL, 64849, México
| | - J F García-Mazcorro
- Research and Development, MNA de México, San Nicolás de los Garza, NL, México
| | - A H Montoya-Rincón
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, NL, México
| | - D L Rodríguez-Reyes
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, NL, México
| | - G Jiménez-Blanco
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, NL, México
| | - M T Alanís Rodríguez
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, NL, México
| | - R Pérez-Cabeza de Vaca
- Coordinación de Investigación y División de Investigación Biomédica, C.M.N. "20 de Noviembre", ISSSTE, Ciudad de México, México
| | - M R Alcorta-García
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, NL, México
- Departamento de Neonatología, Hospital Regional Materno Infantil, Servicios de Salud de Nuevo León, Guadalupe, México
| | - M Brunck
- Tecnológico de Monterrey, Escuela de Ingeniería y Ciencias, Ave. Eugenio Garza Sada 2501 sur, Monterrey, NL, 64849, México
- Division of Experimental Medicine, The Institute for Obesity Research, Tecnológico de Monterrey, Monterrey, NL, México
| | - V J Lara-Díaz
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, NL, México
| | - C Licona-Cassani
- Tecnológico de Monterrey, Escuela de Ingeniería y Ciencias, Ave. Eugenio Garza Sada 2501 sur, Monterrey, NL, 64849, México.
- Division of Integrative Biology, The Institute for Obesity Research, Tecnológico de Monterrey, Monterrey, NL, México.
| |
Collapse
|
28
|
Zhao Y, Qi C, Li X, Lu M, Zhang H, Zhou J, Dang H, Chen J, Li S, Sun J, Yu R, Li D. Prevention of Atopic Dermatitis in Mice by Lactobacillus Reuteri Fn041 Through Induction of Regulatory T Cells and Modulation of the Gut Microbiota. Mol Nutr Food Res 2021; 66:e2100699. [PMID: 34825773 DOI: 10.1002/mnfr.202100699] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 11/11/2021] [Indexed: 11/11/2022]
Abstract
SCOPE The development of atopic dermatitis (AD) in infants is closely related to the lagging development of intestinal microbiota, including that inoculated by breast milk bacteria, and immune development. Lactobacillus reuteri Fn041 is a secretory immunoglobulin A (sIgA) -coated bacterium derived from human milk. METHODS AND RESULTS We intervened with L. reuteri Fn041 in maternal and offspring BALB/C mice during late gestation and lactation and after weaning of the pups, respectively. AD was then induced with MC903. L. reuteri Fn041 significantly suppressed AD symptoms such as skin swelling, mast cell and eosinophil infiltration. This effect was attributed to the regulation of the systemic Th1 and Th2 cytokine ratios and the promotion of CD4+ CD25+ Foxp3+ regulatory T cell proliferation in mesenteric lymph nodes. It is also associated with the regulation of intestinal microbiota, particularly promoting Lactobacillus and Akkermansia. CONCLUSIONS Our study strengthens the understanding that breast milk-derived sIgA coated potential probiotics are involved in the development of infant intestinal microbiota, thus promoting immune development and preventing allergic diseases, and expanding the knowledge of breast milk sIgA and bacterial interactions on infant immune development. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Yuning Zhao
- Institute of Nutrition and Health, Qingdao University, Qingdao, 266071, China
| | - Ce Qi
- Institute of Nutrition and Health, Qingdao University, Qingdao, 266071, China
| | - Xinyue Li
- Institute of Nutrition and Health, Qingdao University, Qingdao, 266071, China
| | - Mengyao Lu
- Institute of Nutrition and Health, Qingdao University, Qingdao, 266071, China
| | - Haowen Zhang
- Institute of Nutrition and Health, Qingdao University, Qingdao, 266071, China
| | - Jingbo Zhou
- Institute of Nutrition and Health, Qingdao University, Qingdao, 266071, China
| | - Hongyang Dang
- Institute of Nutrition and Health, Qingdao University, Qingdao, 266071, China
| | - Jie Chen
- Department of Pediatric Cardiology Nephrology and Rheumatism, The Affiliated, Hospital of Qingdao University Medical College, Qingdao, 266003, China
| | - Shuangqi Li
- Guangzhou Fine Nutrition Research Center, Guangzhou, 510700, China
| | - Jin Sun
- Institute of Nutrition and Health, Qingdao University, Qingdao, 266071, China
| | - Renqiang Yu
- Department of Neonatology, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, 214002, China
| | - Duo Li
- Institute of Nutrition and Health, Qingdao University, Qingdao, 266071, China
| |
Collapse
|
29
|
Abstract
Mother's own milk provides personalized nutrition and immune protection to the developing infant. The presence of healthy microbes plays an important role in the infant's gut by programming the microbiota and excluding potential pathogens. This review describes the important components in mother's own milk that contribute to its superiority for infant nutrition and suggest potential strategies to replicate these factors in alternative feedings when sufficient milk is unavailable. Current strategies to supplement, substitute and replicate mother's own milk including microbial restoration, use of unpasteurized donor human milk, probiotics and fortification are discussed. Critical work remains to be done in understanding the human milk microbiome and metabolome and in improving lactation support for mothers of preterm infants. Increasing delivery of mother's own milk and milk components to infants would likely positively impact infant mortality and health worldwide.
Collapse
Affiliation(s)
- Evon DeBose-Scarlett
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, 2033 Mowry Road Rm 307, Gainesville, FL 32610 USA.
| | - Marion M Bendixen
- College of Nursing, University of Florida, PO Box 100197, Gainesville, FL 32610-0197 USA.
| | - Graciela L Lorca
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, 2033 Mowry Road Rm 307, Gainesville, FL 32610 USA.
| | - Leslie Ann Parker
- College of Nursing, University of Florida, PO Box 100197, Gainesville, FL 32610-0197 USA.
| |
Collapse
|
30
|
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
|
31
|
Olshan KL, Zomorrodi AR, Pujolassos M, Troisi J, Khan N, Fanelli B, Kenyon V, Fasano A, Leonard MM. Microbiota and Metabolomic Patterns in the Breast Milk of Subjects with Celiac Disease on a Gluten-Free Diet. Nutrients 2021; 13:nu13072243. [PMID: 34210038 PMCID: PMC8308312 DOI: 10.3390/nu13072243] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/14/2021] [Accepted: 06/25/2021] [Indexed: 12/30/2022] Open
Abstract
The intestinal microbiome may trigger celiac disease (CD) in individuals with a genetic disposition when exposed to dietary gluten. Research demonstrates that nutrition during infancy is crucial to the intestinal microbiome engraftment. Very few studies to date have focused on the breast milk composition of subjects with a history of CD on a gluten-free diet. Here, we utilize a multi-omics approach with shotgun metagenomics to analyze the breast milk microbiome integrated with metabolome profiling of 36 subjects, 20 with CD on a gluten-free diet and 16 healthy controls. These analyses identified significant differences in bacterial and viral species/strains and functional pathways but no difference in metabolite abundance. Specifically, three bacterial strains with increased abundance were identified in subjects with CD on a gluten-free diet of which one (Rothia mucilaginosa) has been previously linked to autoimmune conditions. We also identified five pathways with increased abundance in subjects with CD on a gluten-free diet. We additionally found four bacterial and two viral species/strains with increased abundance in healthy controls. Overall, the differences observed in bacterial and viral species/strains and in functional pathways observed in our analysis may influence microbiome engraftment in neonates, which may impact their future clinical outcomes.
Collapse
Affiliation(s)
- Katherine L. Olshan
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School, Boston, MA 02114, USA; (K.L.O.); (A.R.Z.); (A.F.)
- Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Boston, MA 02129, USA;
- Department of Pediatrics, Harvard Medical School, Harvard University, Boston, MA 02115, USA
- Celiac Research Program, Harvard Medical School, Boston, MA 02115, USA
| | - Ali R. Zomorrodi
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School, Boston, MA 02114, USA; (K.L.O.); (A.R.Z.); (A.F.)
- Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Boston, MA 02129, USA;
- Department of Pediatrics, Harvard Medical School, Harvard University, Boston, MA 02115, USA
- Celiac Research Program, Harvard Medical School, Boston, MA 02115, USA
| | | | - Jacopo Troisi
- Theoreo srl, University of Salerno, 84084 Salerno, Italy; (M.P.); (J.T.)
- Department of Medicine, Surgery and Dentistry, Scuola Medica Salernitana, University of Salerno, 84084 Salerno, Italy
- European Biomedical Research Institute of Salerno (EBRIS), Via S. De Renzi, 50, 84125 Salerno, Italy
| | - Nayeim Khan
- CosmosID Inc., Rockville, MD 20850, USA; (N.K.); (B.F.)
| | - Brian Fanelli
- CosmosID Inc., Rockville, MD 20850, USA; (N.K.); (B.F.)
| | - Victoria Kenyon
- Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Boston, MA 02129, USA;
- Celiac Research Program, Harvard Medical School, Boston, MA 02115, USA
| | - Alessio Fasano
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School, Boston, MA 02114, USA; (K.L.O.); (A.R.Z.); (A.F.)
- Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Boston, MA 02129, USA;
- Department of Pediatrics, Harvard Medical School, Harvard University, Boston, MA 02115, USA
- Celiac Research Program, Harvard Medical School, Boston, MA 02115, USA
- Department of Medicine, Surgery and Dentistry, Scuola Medica Salernitana, University of Salerno, 84084 Salerno, Italy
| | - Maureen M. Leonard
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School, Boston, MA 02114, USA; (K.L.O.); (A.R.Z.); (A.F.)
- Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Boston, MA 02129, USA;
- Department of Pediatrics, Harvard Medical School, Harvard University, Boston, MA 02115, USA
- Celiac Research Program, Harvard Medical School, Boston, MA 02115, USA
- Correspondence:
| |
Collapse
|
32
|
Lopez Leyva L, Gonzalez E, Li C, Ajeeb T, Solomons NW, Agellon LB, Scott ME, Koski KG. Human Milk Microbiota in an Indigenous Population Is Associated with Maternal Factors, Stage of Lactation, and Breastfeeding Practices. Curr Dev Nutr 2021; 5:nzab013. [PMID: 33898919 PMCID: PMC8053399 DOI: 10.1093/cdn/nzab013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 01/29/2021] [Accepted: 02/17/2021] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Human milk contains a diverse community of bacteria that are modified by maternal factors, but whether these or other factors are similar in developing countries has not been explored. Our objective was to determine whether the milk microbiota was modified by maternal age, BMI, parity, lactation stage, subclinical mastitis (SCM), and breastfeeding practices in the first 6 mo of lactation in an indigenous population from Guatemala. METHODS For this cross-sectional study, Mam-Mayan indigenous mothers nursing infants aged <6 mo were recruited. Unilateral human milk samples were collected (n = 86) and processed for 16S rRNA sequencing at the genus level. Microbial diversity and relative abundance were compared with maternal factors [age, BMI, parity, stage of lactation, SCM, and 3 breastfeeding practices (exclusive, predominant, mixed)] obtained through questionnaires. RESULTS Streptococcus was the most abundant genus (33.8%), followed by Pseudomonas (18.7%) and Sphingobium (10.7%) but relative abundance was associated with maternal factors. First, Lactobacillus and Streptococcus were more abundant in early lactation whereas the common oral (Leptotrichia) and environmental (Comamonas) bacteria were more abundant in established lactation. Second, Streptococcus,Lactobacillus,Lactococcus,Leuconostoc, and Micrococcus had a higher abundance in multiparous mothers compared with primiparous mothers. Third, a more diverse microbiota characterized by a higher abundance of lactic acid bacteria (Lactobacillus,Leuconostoc, and Lactococcus), Leucobacter, and Micrococcus was found in mothers with a healthy BMI. Finally, distinct microbial communities differed by stage of lactation and by exclusive, predominant, or mixed breastfeeding practices. CONCLUSION Milk bacterial communities in an indigenous community were associated with maternal factors. Higher microbial diversity was supported by having a healthy BMI, the absence of SCM, and by breastfeeding. Interestingly, breastfeeding practices when assessed by lactation stage were associated with distinct microbiota profiles.
Collapse
Affiliation(s)
- Lilian Lopez Leyva
- School of Human Nutrition, McGill University, Ste-Anne-de-Bellevue, QC, Canada
| | - Emmanuel Gonzalez
- Canadian Centre for Computational Genomics (C3G), Department of Human Genetics, McGill University and Genome Quebec Innovation Centre, Montréal, QC, Canada
- Microbiome Research Platform, McGill Interdisciplinary Initiative in Infection and Immunity (MI4), Genome Centre, McGill University, Montréal, QC, Canada
| | - Chen Li
- School of Human Nutrition, McGill University, Ste-Anne-de-Bellevue, QC, Canada
| | - Tamara Ajeeb
- School of Human Nutrition, McGill University, Ste-Anne-de-Bellevue, QC, Canada
| | - Noel W Solomons
- Center for Studies of Sensory Impairment, Aging and Metabolism (CeSSIAM), Guatemala City, Guatemala
| | - Luis B Agellon
- School of Human Nutrition, McGill University, Ste-Anne-de-Bellevue, QC, Canada
| | - Marilyn E Scott
- Institute of Parasitology, McGill University, Ste-Anne-de-Bellevue, QC, Canada
| | - Kristine G Koski
- School of Human Nutrition, McGill University, Ste-Anne-de-Bellevue, QC, Canada
| |
Collapse
|
33
|
Qi C, Ding M, Li S, Zhou Q, Li D, Yu R, Sun J. Sex-dependent modulation of immune development in mice by secretory IgA-coated Lactobacillus reuteri isolated from breast milk. J Dairy Sci 2021; 104:3863-3875. [PMID: 33612242 DOI: 10.3168/jds.2020-19437] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 12/04/2020] [Indexed: 12/11/2022]
Abstract
Lactobacilli, commonly present in human breast milk, appear to colonize the neonatal gut and provide protection to infants against various infections, thereby promoting immune development. This study examined the potential probiotic role of breast milk-derived Lactobacillus reuteri FN041 in immune development in mice. The FN041 were gavaged either to BALB/c dams (n = 6/group) during the lactation period or to their offspring (n = 6/sex per intervention) after weaning separately (cointervention). All interventions induced increased intestinal barriers in 5-wk-old offspring, especially in the females. Immunoglobulin A plasmocytes in ileal tissue and secretory IgA (sIgA) in ileal contents increased in all 5-wk-old offspring of cointervention. The activation of mRNA expression of 17 genes was sex-dependent, especially in 5-wk-old offspring. Broader genes were regulated in female mice. The effect of cointervention on the Shannon index of total microbiota is sex-related. The Shannon index of sIgA-coated microbiota increased in both sexes. The sIgA-coated microbiota showed intergroup differences according to β diversity, especially in female mice that showed an increase in Bifidobacterium of Actinobacteria. The sIgA-coated Bifidobacterium was positively correlated with mRNA expression of Tlr9. The sIgA-coated Lactobacillus in male offspring was negatively correlated with mRNA expression of Cldn2. In conclusion, L. reuteri FN041 promoted the production of intestinal sIgA and the expression of genes related to antimicrobial peptides in the offspring and enhanced the function of the mucosal barrier, depending on sex and treatment manner.
Collapse
Affiliation(s)
- Ce Qi
- Institute of Nutrition and Health, Qingdao University, Qingdao 266071, PR China
| | - Mengfan Ding
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China
| | - Shuangqi Li
- Guangzhou Fine Nutrition Research Center, Guangzhou, 510700, PR China
| | - Qin Zhou
- Department of Neonatology, The Affiliated Wuxi Maturity and Child Health Care Hospital of Nanjing Medical University, Wuxi 214002, PR China
| | - Duo Li
- Institute of Nutrition and Health, Qingdao University, Qingdao 266071, PR China
| | - Renqiang Yu
- Department of Neonatology, The Affiliated Wuxi Maturity and Child Health Care Hospital of Nanjing Medical University, Wuxi 214002, PR China.
| | - Jin Sun
- Institute of Nutrition and Health, Qingdao University, Qingdao 266071, PR China.
| |
Collapse
|
34
|
Ding M, Yang B, Ross RP, Stanton C, Zhao J, Zhang H, Chen W. Crosstalk between sIgA-Coated Bacteria in Infant Gut and Early-Life Health. Trends Microbiol 2021; 29:725-735. [PMID: 33602613 DOI: 10.1016/j.tim.2021.01.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 01/24/2021] [Accepted: 01/25/2021] [Indexed: 12/11/2022]
Abstract
Gut microbiota transmission from mother to offspring has attracted much interest in recent years. The gut microbiota in the infant plays a potentially significant role in modulating and maintaining the development of infant immunity. Secretory immunoglobulin A (sIgA), the major immunoglobulin in the intestine, can target polysaccharides and flagellin on the bacterial surface, resulting in sIgA-coated bacteria. The presentation of specific bacteria coated with sIgA may be a signal of disease and provide novel insights into the relationship between infant microbiota and disease. Here, we review the composition of sIgA-coated bacteria in the adult intestine, human milk, and the infant intestine, as well as the factors that influence the development of gut microbiota in early life. Then, we highlight the diseases that are related to variations in sIgA-coated bacteria in the infant and adult intestine. Furthermore, we discuss the possibility that sIgA-coated bacteria could play a role in mediating both innate and adaptive immune responses. Finally, we propose directions for future research to promote our understanding within this field.
Collapse
Affiliation(s)
- Mengfan Ding
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Bo Yang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China; International Joint Research Center for Probiotics and Gut Health, Jiangnan University, Wuxi, Jiangsu, China.
| | - R Paul Ross
- International Joint Research Center for Probiotics and Gut Health, Jiangnan University, Wuxi, Jiangsu, China; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Catherine Stanton
- International Joint Research Center for Probiotics and Gut Health, Jiangnan University, Wuxi, Jiangsu, China; APC Microbiome Ireland, University College Cork, Cork, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland.
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China; International Joint Research Center for Probiotics and Gut Health, Jiangnan University, Wuxi, Jiangsu, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China; Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China.
| |
Collapse
|
35
|
Zhang X, Mushajiang S, Luo B, Tian F, Ni Y, Yan W. The Composition and Concordance of Lactobacillus Populations of Infant Gut and the Corresponding Breast-Milk and Maternal Gut. Front Microbiol 2020; 11:597911. [PMID: 33408705 PMCID: PMC7779531 DOI: 10.3389/fmicb.2020.597911] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 11/02/2020] [Indexed: 12/16/2022] Open
Abstract
The maternal gut is the principal source of commensal bacteria in the infant gut during the lactation stage, where breast milk acts as an intermediary for the transfer of potential probiotic bacteria consortia, including Lactobacillus. This study aimed to characterize the bacterial communities in human milk, maternal, and infant feces in a small yet very homogeneous cohort of 25 healthy mother–infant pairs in northwestern China (n = 25, infant age from 7 days to 2 years), with special emphasis on the cooccurrence and vertical transfer of Lactobacillus phylotypes at the species or strain level in mother-breast milk-infant triads. Accurate sequencing analysis revealed that among 73 Lactobacillus zero-radius operational classification units (ZOTUs) identified, 58 belonging to 18 recognized species or species groups were distributed in all three types of samples. Lactobacillus ruminis, L. mucosae and L. gasseri-johnsonii as true residents were the most represented in all three ecosystems, whereas the content of Lactobacillus phylotypes commonly developed as probiotics was not dominant. While the numbers of Lactobacillus species in breast milk and infant feces were greater than that in maternal feces, principal coordinates analysis (PCoA) based on beta diversity, coupled with the frequency of isolates determined by culture methods, showed that the Lactobacillus community in the infant gut was more similar to that in the maternal gut than to that in breast milk, suggesting that the gut is niche selective for Lactobacillus populations. In addition, identical strains of L. ruminis, L. paracasei, L. mucosae and L. salivarius were isolated from multiple mother–infant pairs, supporting the hypothesis that vertical transfer of bacteria via breastfeeding contributes to the initial establishment of the microbiota in the developing infant intestine.
Collapse
Affiliation(s)
- Xuyao Zhang
- School of Food Science and Technology, Shihezi University, Shihezi, China
| | | | - Baolong Luo
- School of Food Science and Technology, Shihezi University, Shihezi, China
| | - Fengwei Tian
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Yongqing Ni
- School of Food Science and Technology, Shihezi University, Shihezi, China
| | - Wenli Yan
- School of Food Science and Technology, Shihezi University, Shihezi, China
| |
Collapse
|
36
|
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
|
37
|
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: 62] [Impact Index Per Article: 15.5] [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
|
38
|
Changes in the Bacterial Diversity of Human Milk during Late Lactation Period (Weeks 21 to 48). Foods 2020; 9:foods9091184. [PMID: 32867028 PMCID: PMC7554819 DOI: 10.3390/foods9091184] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/24/2020] [Accepted: 08/25/2020] [Indexed: 12/28/2022] Open
Abstract
Breast milk from a single mother was collected during a 28-week lactation period. Bacterial diversity was studied by amplicon sequencing analysis of the V3-V4 variable region of the 16S rRNA gene. Firmicutes and Proteobacteria were the main phyla detected in the milk samples, followed by Actinobacteria and Bacteroidetes. The proportion of Firmicutes to Proteobacteria changed considerably depending on the sampling week. A total of 411 genera or higher taxons were detected in the set of samples. Genus Streptococcus was detected during the 28-week sampling period, at relative abundances between 2.0% and 68.8%, and it was the most abundant group in 14 of the samples. Carnobacterium and Lactobacillus had low relative abundances. At the genus level, bacterial diversity changed considerably at certain weeks within the studied period. The weeks or periods with lowest relative abundance of Streptococcus had more diverse bacterial compositions including genera belonging to Proteobacteria that were poorly represented in the rest of the samples.
Collapse
|
39
|
The Infant Gut Microbiota and Risk of Asthma: The Effect of Maternal Nutrition during Pregnancy and Lactation. Microorganisms 2020; 8:microorganisms8081119. [PMID: 32722458 PMCID: PMC7466123 DOI: 10.3390/microorganisms8081119] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 02/07/2023] Open
Abstract
Research has amply demonstrated that early life dysbiosis of the gut microbiota influences the propensity to develop asthma. The influence of maternal nutrition on infant gut microbiota is therefore of growing interest. However, a handful of prospective studies have examined the role of maternal dietary patterns during pregnancy in influencing the infant gut microbiota but did not assess whether this resulted in an increased risk of asthma later in life. The mechanisms involved in the process are also, thus far, poorly documented. There have also been few studies examining the effect of maternal dietary nutrient intake during lactation on the milk microbiota, the effect on the infant gut microbiota and, furthermore, the consequences for asthma development remain largely unknown. Therefore, the specific aim of this mini review is summarizing the current knowledge regarding the effect of maternal nutrition during pregnancy and lactation on the infant gut microbiota composition, and whether it has implications for asthma development.
Collapse
|
40
|
Zimmermann P, Curtis N. Breast milk microbiota: A review of the factors that influence composition. J Infect 2020; 81:17-47. [PMID: 32035939 DOI: 10.1016/j.jinf.2020.01.023] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/29/2020] [Accepted: 01/30/2020] [Indexed: 01/31/2023]
Abstract
Breastfeeding is associated with considerable health benefits for infants. Aside from essential nutrients, immune cells and bioactive components, breast milk also contains a diverse range of microbes, which are important for maintaining mammary and infant health. In this review, we summarise studies that have investigated the composition of the breast milk microbiota and factors that might influence it. We identified 44 studies investigating 3105 breast milk samples from 2655 women. Several studies reported that the bacterial diversity is higher in breast milk than infant or maternal faeces. The maximum number of each bacterial taxonomic level detected per study was 58 phyla, 133 classes, 263 orders, 596 families, 590 genera, 1300 species and 3563 operational taxonomic units. Furthermore, fungal, archaeal, eukaryotic and viral DNA was also detected. The most frequently found genera were Staphylococcus, Streptococcus Lactobacillus, Pseudomonas, Bifidobacterium, Corynebacterium, Enterococcus, Acinetobacter, Rothia, Cutibacterium, Veillonella and Bacteroides. There was some evidence that gestational age, delivery mode, biological sex, parity, intrapartum antibiotics, lactation stage, diet, BMI, composition of breast milk, HIV infection, geographic location and collection/feeding method influence the composition of the breast milk microbiota. However, many studies were small and findings sometimes contradictory. Manipulating the microbiota by adding probiotics to breast milk or artificial milk offers an exciting avenue for future interventions to improve infant health.
Collapse
Affiliation(s)
- Petra Zimmermann
- Department of Paediatrics, Fribourg Hospital HFR and Faculty of Science and Medicine, University of Fribourg, Switzerland; Department of Paediatrics, The University of Melbourne, Parkville, Australia; Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, Australia; Infectious Diseases Unit, The Royal Children's Hospital Melbourne, Parkville, Australia.
| | - Nigel Curtis
- Department of Paediatrics, The University of Melbourne, Parkville, Australia; Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, Australia; Infectious Diseases Unit, The Royal Children's Hospital Melbourne, Parkville, Australia
| |
Collapse
|
41
|
Guo WL, Chen M, Pan WL, Zhang Q, Xu JX, Lin YC, Li L, Liu B, Bai WD, Zhang YY, Ni L, Rao PF, Lv XC. Hypoglycemic and hypolipidemic mechanism of organic chromium derived from chelation of Grifola frondosa polysaccharide-chromium (III) and its modulation of intestinal microflora in high fat-diet and STZ-induced diabetic mice. Int J Biol Macromol 2020; 145:1208-1218. [DOI: 10.1016/j.ijbiomac.2019.09.206] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 09/23/2019] [Accepted: 09/26/2019] [Indexed: 02/06/2023]
|
42
|
Douglas CA, Ivey KL, Papanicolas LE, Best KP, Muhlhausler BS, Rogers GB. DNA extraction approaches substantially influence the assessment of the human breast milk microbiome. Sci Rep 2020; 10:123. [PMID: 31924794 PMCID: PMC6954186 DOI: 10.1038/s41598-019-55568-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 11/20/2019] [Indexed: 01/21/2023] Open
Abstract
In addition to providing nutritional and bioactive factors necessary for infant development, human breast milk contains bacteria that contribute to the establishment of commensal microbiota in the infant. However, the composition of this bacterial community differs considerably between studies. We hypothesised that bacterial DNA extraction methodology from breast milk samples are a substantial contributor to these inter-study differences. We tested this hypothesis by applying five widely employed methodologies to a mock breast milk sample and four individual human breast milk samples. Significant differences in DNA yield and purity were observed between methods (P < 0.05). Microbiota composition, assessed by 16S rRNA gene amplicon sequencing, also differed significantly with extraction methodology (P < 0.05), including in the contribution of contaminant signal. Concerningly, many of the bacterial taxa identified here as contaminants have been reported as components of the breast milk microbiome in other studies. These findings highlight the importance of using stringent, well-validated, DNA extraction methodologies for analysis of the breast milk microbiome, and exercising caution interpreting microbiota data from low-biomass contexts.
Collapse
Affiliation(s)
- Chloe A Douglas
- South Australian Health and Medical Research Institute, Healthy Mothers, Babies and Children Theme, Women's and Children's Hospital, King William Road, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Microbiome & Host Health Programme, Adelaide, South Australia, Australia
- Faculty of Health Science, Discipline of Medicine - Paediatrics and Reproductive Health, The University of Adelaide, Adelaide, South Australia, Australia
| | - Kerry L Ivey
- South Australian Health and Medical Research Institute, Microbiome & Host Health Programme, Adelaide, South Australia, Australia
- Harvard T.H. Chan School of Public Health, Department of Nutrition, Boston, Massachusetts, USA
| | - Lito E Papanicolas
- South Australian Health and Medical Research Institute, Microbiome & Host Health Programme, Adelaide, South Australia, Australia
| | - Karen P Best
- South Australian Health and Medical Research Institute, Healthy Mothers, Babies and Children Theme, Women's and Children's Hospital, King William Road, Adelaide, South Australia, Australia
| | - Beverly S Muhlhausler
- South Australian Health and Medical Research Institute, Healthy Mothers, Babies and Children Theme, Women's and Children's Hospital, King William Road, Adelaide, South Australia, Australia
- Nutrition & Health Program, Health and Biosecurity, CSIRO, Adelaide, South Australia, Australia
- Department of Food and Wine Science, School of Agriculture, Food and Wine, The University of Adelaide, Adelaide, South Australia, Australia
| | - Geraint B Rogers
- South Australian Health and Medical Research Institute, Microbiome & Host Health Programme, Adelaide, South Australia, Australia.
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia.
| |
Collapse
|
43
|
Ge Y, Lin S, Li B, Yang Y, Tang X, Shi Y, Sun J, Le G. Oxidized Pork Induces Oxidative Stress and Inflammation by Altering Gut Microbiota in Mice. Mol Nutr Food Res 2019; 64:e1901012. [PMID: 31845486 DOI: 10.1002/mnfr.201901012] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/26/2019] [Indexed: 12/14/2022]
Abstract
SCOPE Reduced digestibility of foods containing oxidized proteins and the subsequent excessive accumulation of undigested components in the colon may cause changes in the intestinal flora composition. This study evaluates the characteristics of this change and the potential adverse effects on organisms. METHODS AND RESULTS Pork is cooked using sous-vide or at high temperature and pressure (HTP), then freeze-dried, resulting in different levels of oxidized damage. Mice are fed diets containing low- (LOP), medium- (MOP), or high-oxidative damage pork (HOP) for 12 weeks. HOP intake increases mice body weight, induces inflammatory response, and causes oxidative stress, as indicated by the accumulation of oxidative products. Increased serum LPS levels and downregulation of tight junction-related genes in the mucosa suggest mucosal barrier damage. Alterations in the cecal microbiota include reduced relative abundance of the mucin-degrading bacteria Akkermansia, beneficial bacteria Lactobacillus and Bifidobacterium, and H2 S-producing bacteria Desulfovibrio and increased relative abundance of the pro-inflammatory bacteria Escherichia-Shigella and pathobiont Mucispirillum. CONCLUSION HOP intake causes the accumulation of oxidative products, increases body weight, damages the intestinal barrier, and induces oxidative stress and inflammatory response, likely by altering gut microbiota through protein oxidation (POX).
Collapse
Affiliation(s)
- Yueting Ge
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China.,Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| | - Shiman Lin
- Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| | - Bowen Li
- Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| | - Yuhui Yang
- Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China.,College of Grain and Food Science, Henan University of Technology, Zhengzhou, 450001, P. R. China
| | - Xue Tang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China.,Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| | - Yonghui Shi
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China.,Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| | - Jin Sun
- Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China.,Institute of Nutrition and Health, Qingdao University, Qingdao, 266071, P. R. China
| | - Guowei Le
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China.,Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| |
Collapse
|
44
|
Guo WL, Deng JC, Pan YY, Xu JX, Hong JL, Shi FF, Liu GL, Qian M, Bai WD, Zhang W, Liu B, Zhang YY, Luo PJ, Ni L, Rao PF, Lv XC. Hypoglycemic and hypolipidemic activities of Grifola frondosa polysaccharides and their relationships with the modulation of intestinal microflora in diabetic mice induced by high-fat diet and streptozotocin. Int J Biol Macromol 2019; 153:1231-1240. [PMID: 31759027 DOI: 10.1016/j.ijbiomac.2019.10.253] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 10/25/2019] [Accepted: 10/28/2019] [Indexed: 02/07/2023]
Abstract
This study aimed to investigate the hypoglycemic and hypolipidemic activities of polysaccharides from Grifola frondosa (GFP) in diabetic mice induced by high-fat diet (HFD) and streptozotocin (STZ). Results showed that oral administration of GFP markedly reduced the serum levels of fasting blood glucose (FBG), oral glucose tolerance (OGT), cholesterol (TC), triglyceride (TG) and low-density lipoprotein cholesterol (LDL-C), and significantly decreased the hepatic levels of TC, TG and free fatty acids (FFA). Meanwhile, high-dose of GFP supplementation (900 mg/kg day) also showed powerful effects on moderating the composition of intestinal microflora in diabetic mice, especially altering the functionally relevant intestinal microbial phylotypes. Spearman's correlation network analysis revealed that key microbial phylotypes responding to GFP intervention were strongly correlated with the glucose and lipid metabolic disorders associated parameters. Moreover, GFP treatment regulated mRNA expression levels of the genes responsible for hepatic glucose and lipid metabolism. It is noteworthy that GFP treatment markedly increased mRNA expression of cholesterol 7α-hydroxylase (CYP7A1) and bile salt export pump (BSEP), suggesting an enhancement of bile acids (BAs) synthesis and excretion in liver. These findings demonstrated that GFP could prevent hyperglycemia and hyperlipidemia in diabetic mice by altering gut microbiota and regulating hepatic glycolipid metabolism related genes, and therefore could be used as potential functional food ingredients for the prevention or treatment of hyperglycemia and hyperlipidemia.
Collapse
Affiliation(s)
- Wei-Ling Guo
- Institute of Food Science and Technology, College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China; National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Jia-Cong Deng
- School of Ocean Science and Biochemistry Engineering, Fuqing Branch of Fujian Normal University, Fuqing, Fujian 350300, China
| | - Yu-Yang Pan
- National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Jia-Xin Xu
- National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Jia-Li Hong
- National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Fei-Fei Shi
- Institute of Food Science and Technology, College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China; National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Gong-Liang Liu
- College of Light Industry and Food Science, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Min Qian
- College of Light Industry and Food Science, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Wei-Dong Bai
- College of Light Industry and Food Science, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Wen Zhang
- Institute of Food Science and Technology, College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Bin Liu
- National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Yan-Yan Zhang
- Institute of Food Science and Technology, College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China; Department of Flavor Chemistry, Institute of Food Science and Biotechnology, University of Hohenheim, Stuttgart 70599, Germany
| | - Peng-Jie Luo
- China National Center for Food Safety Risk Assessment, NHC Key Laboratory of Food Safety Risk Assessment, Beijing 100022, China.
| | - Li Ni
- Institute of Food Science and Technology, College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Ping-Fan Rao
- Institute of Food Science and Technology, College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Xu-Cong Lv
- Institute of Food Science and Technology, College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China; National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China; Department of Flavor Chemistry, Institute of Food Science and Biotechnology, University of Hohenheim, Stuttgart 70599, Germany.
| |
Collapse
|
45
|
Cao X, Zheng Y, Wu S, Yang N, Wu J, Liu B, Ye W, Yang M, Yue X. Characterization and comparison of milk fat globule membrane N-glycoproteomes from human and bovine colostrum and mature milk. Food Funct 2019; 10:5046-5058. [PMID: 31359016 DOI: 10.1039/c9fo00686a] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Human and bovine milk fat globule membrane (MFGM) proteins have been identified and characterized; however, their glycosylation during lactation remains unclear. We adopted a glycoproteomics approach to profile and compare MFGM N-glycoproteomes in human and bovine milk during lactation. A total of 843, 718, 614, and 273 N-glycosite peptides corresponding to 465, 423, 334, and 176 glycoproteins were identified in human colostrum, human mature milk, bovine colostrum, and bovine mature milk, respectively. The biological functions of these MFGM N-glycoproteins were revealed through bioinformatics. Substantial differences were observed between human and bovine milk, and immune-related MFGM N-glycoproteins varied between colostrum and mature milk from both species. Our results expand current knowledge of MFGM N-glycoproteomes, and further demonstrate the complexity and biological functions of MFGM N-glycosylation. These data can provide references for the application of bovine MFGM N-glycoproteins in infant formula to resemble human milk and in functional foods.
Collapse
Affiliation(s)
- Xueyan Cao
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, PR China.
| | | | | | | | | | | | | | | | | |
Collapse
|