1
|
Patel M, Patel K. Emerging insights of Staphylococcus spp. in human mastitis. Microb Pathog 2025; 205:107685. [PMID: 40349998 DOI: 10.1016/j.micpath.2025.107685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 05/01/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
Human mastitis represents a prevalent and intricate condition that significantly challenges breastfeeding women, often exacerbated by pathogenic bacteria such as Staphylococcus aureus. A deep understanding of the interplay between human mastitis, the breast milk microbiome, and causative agents is imperative. This understanding must focus on the bacterium's virulence and resistance genes, which critically influence the severity and persistence of mastitis. Current methods for detecting these genes, including Polymerase Chain Reaction (PCR), 16S rRNA gene sequencing, shotgun metagenomic sequencing, multiplex PCR, whole genome sequencing (WGS), loop-mediated isothermal amplification (LAMP), CRISPR-based assays, and microarray technology, are vital in elucidating bacterial pathogenicity and resistance profiles. However, advanced attention is required to refine diagnostic techniques, enabling earlier detection and more effective therapeutic approaches for human mastitis. The involvement of Staphylococcus aureus in human infection should be a prime focus, especially in women's health, which deals directly with neonates. Essential virulence genes in Staphylococcus species are instrumental in infection mechanisms and antibiotic resistance, serving as potential targets for personalized treatments. Thus, this review focuses on Staphylococcusaureus-induced mastitis, examining its virulence factors and detection techniques to advance diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Mansi Patel
- Department of Medical Laboratory Technology, Bapubhai Desaibhai Patel Institute of Paramedical Sciences (BDIPS), Charotar University of Science and Technology (CHARUSAT), Anand, Gujarat, 388421, India
| | - Khushal Patel
- Department of Medical Laboratory Technology, Bapubhai Desaibhai Patel Institute of Paramedical Sciences (BDIPS), Charotar University of Science and Technology (CHARUSAT), Anand, Gujarat, 388421, India.
| |
Collapse
|
2
|
Ge H, Zhu W, Zhang J, Wang Z, Shi H, Sun J, Shi M. Human milk microbiota and oligosaccharides in colostrum and mature milk: comparison and correlation. Front Nutr 2024; 11:1512700. [PMID: 39726867 PMCID: PMC11670000 DOI: 10.3389/fnut.2024.1512700] [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: 10/17/2024] [Accepted: 11/21/2024] [Indexed: 12/28/2024] Open
Abstract
Background The interaction between the human breast milk microbiota and human milk oligosaccharides (HMOs) plays a crucial role in the healthy growth and development of infants. We aimed to clarify the link between the breast milk microbiota and HMOs at two stages of lactation. Methods The microbiota and HMOs of 20 colostrum samples (C group, 1-5 days postpartum) and 20 mature milk samples (S group, 42 days postpartum) collected from postpartum mothers were analyzed using 16S rRNA gene high-throughput sequencing and high-performance liquid chromatography-tandem mass spectrometry. Result The total average HMO content was significantly higher in the C group than in the S group (6.76 ± 1.40 g/L vs. 10.27 ± 2.00 g/L, p < 0.05). Among the HMOs, the average values of 2'-fucosyllactose (2'-FL, 1.64 ± 1.54 g/L vs. 3.03 ± 1.79 g/L), 3'-sialyllactose (3'-SL, 0.10 ± 0.02 g/L vs. 0.21 ± 0.06), 6'-SL (0.22 ± 0.09 g/L vs. 0.33 ± 0.11 g/L), and lacto-N-triaose 2 (LNT2, 0.03 ± 0.01 g/L vs. 0.16 ± 0.08 g/L) were significantly lower in the S group than in the C group (p < 0.05), while that of 3'-FL was significantly higher in the S group than in the C group (1.35 ± 1.00 g/L vs. 0.41 ± 0.43 g/L, p < 0.05). The diversity and structure of the microbiota in the S and C groups were also significantly different (p < 0.05). Comparative analysis of the microbial communities revealed that Proteobacteria and Firmicutes were the most abundant phyla, in both groups, with the keystone species (Serratia, Streptococcus and Staphylococcus) of breast milk closely interacting with HMOs, including 3'-SL, 6'-SL, and LNT2. In PICRUSt2 functional prediction analysis, the S group exhibited significant reduction in the expression of genes involved in several infectious disease pathways. Discussion Our findings support the recognition of human milk as a synbiotic comprising beneficial bacteria and prebiotic HMOs.
Collapse
Affiliation(s)
- Hongda Ge
- Department of Clinical Laboratory, Dalian Women and Children’s Medical Group, Dalian, China
| | - Wenxiu Zhu
- Centre for Reproductive and Genetic Medicine, Dalian Women and Children’s Medical Group, Dalian, China
| | - Jing Zhang
- Department of Clinical Laboratory, Central Hospital of Dalian University of Technology, Dalian Municipal Central Hospital, Dalian, China
| | - Zijing Wang
- Maternity Ward, Dalian Women and Children’s Medical Group, Dalian, China
| | - Huijing Shi
- Maternity Ward, Dalian Women and Children’s Medical Group, Dalian, China
| | - Jie Sun
- Child Health Care Clinic, Dalian Women and Children’s Medical Group, Dalian, China
| | - Ming Shi
- Department of Clinical Laboratory, Dalian Women and Children’s Medical Group, Dalian, China
- Centre for Reproductive and Genetic Medicine, Dalian Women and Children’s Medical Group, Dalian, China
| |
Collapse
|
3
|
Vélez-Ixta JM, Juárez-Castelán CJ, Ramírez-Sánchez D, Lázaro-Pérez NDS, Castro-Arellano JJ, Romero-Maldonado S, Rico-Arzate E, Hoyo-Vadillo C, Salgado-Mancilla M, Gómez-Cruz CY, Krishnakumar A, Piña-Escobedo A, Benitez-Guerrero T, Pizano-Zárate ML, Cruz-Narváez Y, García-Mena J. Post Natal Microbial and Metabolite Transmission: The Path from Mother to Infant. Nutrients 2024; 16:1990. [PMID: 38999737 PMCID: PMC11243545 DOI: 10.3390/nu16131990] [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: 06/01/2024] [Revised: 06/20/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
The entero-mammary pathway is a specialized route that selectively translocates bacteria to the newborn's gut, playing a crucial role in neonatal development. Previous studies report shared bacterial and archaeal taxa between human milk and neonatal intestine. However, the functional implications for neonatal development are not fully understood due to limited evidence. This study aimed to identify and characterize the microbiota and metabolome of human milk, mother, and infant stool samples using high-throughput DNA sequencing and FT-ICR MS methodology at delivery and 4 months post-partum. Twenty-one mothers and twenty-five infants were included in this study. Our results on bacterial composition suggest vertical transmission of bacteria through breastfeeding, with major changes occurring during the first 4 months of life. Metabolite chemical characterization sheds light on the growing complexity of the metabolites. Further data integration and network analysis disclosed the interactions between different bacteria and metabolites in the biological system as well as possible unknown pathways. Our findings suggest a shared bacteriome in breastfed mother-neonate pairs, influenced by maternal lifestyle and delivery conditions, serving as probiotic agents in infants for their healthy development. Also, the presence of food biomarkers in infants suggests their origin from breast milk, implying selective vertical transmission of these features.
Collapse
Affiliation(s)
- Juan Manuel Vélez-Ixta
- Departamento de Genética y Biología Molecular, Cinvestav, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico; (J.M.V.-I.); (C.J.J.-C.); (D.R.-S.); (N.d.S.L.-P.); (A.K.); (A.P.-E.); (T.B.-G.)
| | - Carmen Josefina Juárez-Castelán
- Departamento de Genética y Biología Molecular, Cinvestav, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico; (J.M.V.-I.); (C.J.J.-C.); (D.R.-S.); (N.d.S.L.-P.); (A.K.); (A.P.-E.); (T.B.-G.)
| | - Daniela Ramírez-Sánchez
- Departamento de Genética y Biología Molecular, Cinvestav, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico; (J.M.V.-I.); (C.J.J.-C.); (D.R.-S.); (N.d.S.L.-P.); (A.K.); (A.P.-E.); (T.B.-G.)
| | - Noemí del Socorro Lázaro-Pérez
- Departamento de Genética y Biología Molecular, Cinvestav, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico; (J.M.V.-I.); (C.J.J.-C.); (D.R.-S.); (N.d.S.L.-P.); (A.K.); (A.P.-E.); (T.B.-G.)
| | - José Javier Castro-Arellano
- Laboratorio de Posgrado e Investigación de Operaciones Unitarias, Escuela Superior de Ingeniería Química e Industrias Extractivas, Instituto Politécnico Nacional, Mexico City 07738, Mexico; (J.J.C.-A.); (E.R.-A.); (M.S.-M.); (C.Y.G.-C.)
| | - Silvia Romero-Maldonado
- Unidad de Cuidados Intermedios al Recién Nacido, Instituto Nacional de Perinatología, Secretaría de Salud, Mexico City 11000, Mexico;
| | - Enrique Rico-Arzate
- Laboratorio de Posgrado e Investigación de Operaciones Unitarias, Escuela Superior de Ingeniería Química e Industrias Extractivas, Instituto Politécnico Nacional, Mexico City 07738, Mexico; (J.J.C.-A.); (E.R.-A.); (M.S.-M.); (C.Y.G.-C.)
| | - Carlos Hoyo-Vadillo
- Departamento de Farmacología, Cinvestav, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico;
| | - Marisol Salgado-Mancilla
- Laboratorio de Posgrado e Investigación de Operaciones Unitarias, Escuela Superior de Ingeniería Química e Industrias Extractivas, Instituto Politécnico Nacional, Mexico City 07738, Mexico; (J.J.C.-A.); (E.R.-A.); (M.S.-M.); (C.Y.G.-C.)
| | - Carlos Yamel Gómez-Cruz
- Laboratorio de Posgrado e Investigación de Operaciones Unitarias, Escuela Superior de Ingeniería Química e Industrias Extractivas, Instituto Politécnico Nacional, Mexico City 07738, Mexico; (J.J.C.-A.); (E.R.-A.); (M.S.-M.); (C.Y.G.-C.)
| | - Aparna Krishnakumar
- Departamento de Genética y Biología Molecular, Cinvestav, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico; (J.M.V.-I.); (C.J.J.-C.); (D.R.-S.); (N.d.S.L.-P.); (A.K.); (A.P.-E.); (T.B.-G.)
| | - Alberto Piña-Escobedo
- Departamento de Genética y Biología Molecular, Cinvestav, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico; (J.M.V.-I.); (C.J.J.-C.); (D.R.-S.); (N.d.S.L.-P.); (A.K.); (A.P.-E.); (T.B.-G.)
| | - Tizziani Benitez-Guerrero
- Departamento de Genética y Biología Molecular, Cinvestav, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico; (J.M.V.-I.); (C.J.J.-C.); (D.R.-S.); (N.d.S.L.-P.); (A.K.); (A.P.-E.); (T.B.-G.)
| | - María Luisa Pizano-Zárate
- Coordinación de Nutrición y Bioprogramación, Instituto Nacional de Perinatología, Secretaría de Salud, Mexico City 11000, Mexico
- Unidad de Medicina Familiar No. 4, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| | - Yair Cruz-Narváez
- Laboratorio de Posgrado e Investigación de Operaciones Unitarias, Escuela Superior de Ingeniería Química e Industrias Extractivas, Instituto Politécnico Nacional, Mexico City 07738, Mexico; (J.J.C.-A.); (E.R.-A.); (M.S.-M.); (C.Y.G.-C.)
| | - Jaime García-Mena
- Departamento de Genética y Biología Molecular, Cinvestav, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico; (J.M.V.-I.); (C.J.J.-C.); (D.R.-S.); (N.d.S.L.-P.); (A.K.); (A.P.-E.); (T.B.-G.)
| |
Collapse
|
4
|
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
|
5
|
Dinleyici M, Pérez-Brocal V, Arslanoglu S, Aydemir O, Sevuk Ozumut S, Tekin N, Vandenplas Y, Moya A, Dinleyici EC. Composition of Microbiota in Transient and Mature Human Milk: Significant Changes in Large for Gestational Age Group. Nutrients 2024; 16:208. [PMID: 38257101 PMCID: PMC10818272 DOI: 10.3390/nu16020208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/03/2024] [Accepted: 01/06/2024] [Indexed: 01/24/2024] Open
Abstract
The composition of the human milk (HM) microbiota and, consequently, the microorganisms that are passed on to the infant through breastfeeding, can be influenced by various factors such as the mother's health and diet, gestational age, delivery mode, lactation stage, method of infant feeding, and geographical location. The aim of the Human Milk-Gest Study was to compare the microbiota of transient (postpartum 7-15 days) and mature HM (postpartum 45-90 days) of 44 mothers, and to investigate any potential changes associated with preterm birth, mode of delivery, and birth weight in relation to gestational age. The data were classified into five study groups: normal spontaneous delivery-term (NS-T) newborns, cesarean delivery-term (CS-T) newborns, preterm (PT) newborns (with a gestational age of less than 37 weeks), small for gestational age (SGA) newborns, and large for gestational age (LGA) newborns. An analysis of differential abundance was conducted using ANCOM-BC to compare the microbial genera between transient and mature HM samples as well as between other study groups. A significant difference was detected between HM samples at different sampling times and between the study groups (p < 0.01). In transient HM samples, Ralstonia, Burkholderiaceae_uc, and Pelomonas were significantly dominant in the LGA group compared to the NS-T, CS-T, PT, and SGA groups. In mature HM samples, Burkholderiaceae_uc, Ralstonia, Pelomonas, and Klebsiella were significantly dominant in the LGA group compared to the NS-T, CS-T, and PT groups, while Ralstonia, Burkholderiaceae_uc, and Pelomonas were significantly dominant in the LGA group compared to the SGA group. Differences were also detected between the transient and mature HM samples in the CS-T, PT, SGA, and LGA groups, but no differences occurred in the NS-T groups. In conclusion, we showed that Ralstonia, Burkholderiaceae_uc, and Pelomonas were significantly dominant in the LGA group in transient HM and continued in mature HM. The body mass index (BMI) of the mothers in the LGA group was not >30 at conception, however, the maternal BMI at birth and maternal weight gain during pregnancy were higher than in the other groups. The nutritional composition of HM is specifically designed to meet infant nutritional requirements during early life. Evaluating the effects of HM microbiota on infant microbiota composition and short- and long-term health effects in larger studies would be useful.
Collapse
Affiliation(s)
- Meltem Dinleyici
- Department of Social Pediatrics, Faculty of Medicine, Eskisehir Osmangazi University, 26480 Eskisehir, Türkiye;
| | - Vicente Pérez-Brocal
- Department of Genomics and Health, Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO-Public Health), 46020 Valencia, Spain; (V.P.-B.)
- CIBER in Epidemiology and Public Health (CIBEResp), 28029 Madrid, Spain
| | - Sertac Arslanoglu
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Medeniyet University, 34720 Istanbul, Türkiye
| | - Ozge Aydemir
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Eskisehir Osmangazi University, 26040 Eskisehir, Türkiye; (O.A.); (N.T.)
| | - Sibel Sevuk Ozumut
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Medeniyet University, 34720 Istanbul, Türkiye
| | - Neslihan Tekin
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Eskisehir Osmangazi University, 26040 Eskisehir, Türkiye; (O.A.); (N.T.)
| | - Yvan Vandenplas
- KidZ Health Castle, UZ Brussel, Vrije Unversiteit Brussel, 1090 Brussels, Belgium
| | - Andrés Moya
- Department of Genomics and Health, Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO-Public Health), 46020 Valencia, Spain; (V.P.-B.)
- CIBER in Epidemiology and Public Health (CIBEResp), 28029 Madrid, Spain
- Institute for Integrative Systems Biology (I2SysBio), University of Valencia and Spanish National Research Council (CSIC-UVEG), 46980 Valencia, Spain
| | - Ener Cagri Dinleyici
- Department of Pediatrics, Faculty of Medicine, Eskisehir Osmangazi University, 26040 Eskisehir, Türkiye
| |
Collapse
|
6
|
Giovannini N, Lattuada D, Danusso R, Ferrazzi E. From pandemic to syndemic: microbiota, pregnancy, and environment at a crossroad. J Matern Fetal Neonatal Med 2023; 36:2183738. [PMID: 36977591 DOI: 10.1080/14767058.2023.2183738] [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: 03/30/2023]
Abstract
Aim: SARS-CoV2 is the latest pandemic that have plagued the socio-health system as an epiphenomenon resulting from planetary resources abuse, crucial for biodiversity. The Anthropocene best defines the present epoch in which human activity irreversibly manipulates intricate and delicate geological and biological balances established over eons. The devastating ecological and socio-economic implications of COVID-19, underline the importance of updating the present pandemic framework to a syndemic. This paper stems from the need to suggest to scientists, doctors, and patients a mission that integrates responsibility from individual to collective health, from present to trans-generational, from human to the entire biotic network. Today's choices are crucial for the perspective on all levels: political, economic, and health as well as cultural.Methods: Research on PubMed and other specific web-sites journal was performed on the topic "Microbiota", "Covid-19", "Pandemic", "Zoonosis", "SARS-CoV-2", "Environmental Pollutants", "Epigenetics", "Fetal Programming", "Human Extinction". Data collected were analysed for an integrative model of interconnection between environment, pregnancy, SARS-CoV-2 infection, and microbiota. Moreover, systematic literature review allowed to summarise in a table information about the worst pandemics that afflicted the human species recently.Results: This paper offers a broad view of the current pandemic starting with pregnancy, the moment when a new life begins and the health trajectories of the unborn child are defined, which will inevitably have repercussions on his well-being. The fundamental role of the biodiversity-rich microbiota in avoiding the development of severe infectious diseases, is therefore highlighted. It is imperative to adjust the current reductionist paradigm based on mostly immediate symptom management towards a broader understanding of the spatial interconnection of ecological niches with human health and the impacts of today's choices on the future. Health and healthcare are elitist rather than egalitarian, therefore focusing on environmental health forces us to make a concerted and systemic effort that challenges political and economic barriers, which are biologically senseless. A healthy microbiota is essential to well-being, both by preventing chronic degenerative conditions, the infectiousness and pathogenicity of bacterial and viral diseases. SARS-CoV-2 should not be an exception. The human microbiota, forged by the first 1,000 days of life, is fundamental in shaping the health-disease trajectories, and by the everlasting exposome that is dramatically affected by the ecological disaster. Individual health is one world health whereas single and global well-being are interdependent in a space-time perspective.Conclusions: Is it not a convenient reductionism not to consider the COVID-19 emergency as a bio-social epiphenomenon of a far more devastating and multi-faceted crisis whose common denominator is the global biotic network loss of which humans are still part?
Collapse
Affiliation(s)
- Niccolò Giovannini
- Department of women-child-newborn Obstetrics and Gynaecology, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Debora Lattuada
- Department of women-child-newborn Obstetrics and Gynaecology, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Roberta Danusso
- Department of women-child-newborn Obstetrics and Gynaecology, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Enrico Ferrazzi
- Department of women-child-newborn Obstetrics and Gynaecology, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| |
Collapse
|
7
|
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
|
8
|
Spreckels JE, Fernández-Pato A, Kruk M, Kurilshikov A, Garmaeva S, Sinha T, Ghosh H, Harmsen H, Fu J, Gacesa R, Zhernakova A. Analysis of microbial composition and sharing in low-biomass human milk samples: a comparison of DNA isolation and sequencing techniques. ISME COMMUNICATIONS 2023; 3:116. [PMID: 37945978 PMCID: PMC10636111 DOI: 10.1038/s43705-023-00325-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 10/18/2023] [Accepted: 10/26/2023] [Indexed: 11/12/2023]
Abstract
Human milk microbiome studies are currently hindered by low milk bacterial/human cell ratios and often rely on 16S rRNA gene sequencing, which limits downstream analyses. Here, we aimed to find a method to study milk bacteria and assess bacterial sharing between maternal and infant microbiota. We tested four DNA isolation methods, two bacterial enrichment methods and three sequencing methods on mock communities, milk samples and negative controls. Of the four DNA isolation kits, the DNeasy PowerSoil Pro (PS) and MagMAX Total Nucleic Acid Isolation (MX) kits provided consistent 16S rRNA gene sequencing results with low contamination. Neither enrichment method substantially decreased the human metagenomic sequencing read-depth. Long-read 16S-ITS-23S rRNA gene sequencing biased the mock community composition but provided consistent results for milk samples, with little contamination. In contrast to 16S rRNA gene sequencing, 16S-ITS-23S rRNA gene sequencing of milk, infant oral, infant faecal and maternal faecal DNA from 14 mother-infant pairs provided sufficient resolution to detect significantly more frequent sharing of bacteria between related pairs compared to unrelated pairs. In conclusion, PS or MX kit-DNA isolation followed by 16S rRNA gene sequencing reliably characterises human milk microbiota, and 16S-ITS-23S rRNA gene sequencing enables studies of bacterial transmission in low-biomass samples.
Collapse
Grants
- This study was supported by funds from the Dutch Research Council (NWO-VIDI grant 016.178.056 to A.Z., NWO-VICI grant VI.C.202.022 to J.F., NWO gravitation grant Exposome-NL 024.004.017 to A.K. and A.Z., NWO gravitation grant Netherlands Organ-on-Chip Initiative 024.003.001 to J.F.), the Dutch Heart Foundation (IN-CONTROL CVON2018-27 to J.F.), the European Research Council (ERC starting grant 715772 to A.Z., ERC consolidator grant 101001678 to J.F.), an EASI-Genomics grant (PID7780 to T.S. and A.Z.), the De-Cock Hadders foundation (2021-57 to J.E.S., 2021-08 to S.G.), the International Society for Research in Human Milk and Lactation (ISRHML, personal grant to J.E.S), the Winston Bakker Fonds (WB-08, granted to T.S.), and the European Union’s Horizon 2020 research innovation program (824110). S.G. and T.S. hold scholarships from the Graduate School of Medical Sciences and the Junior Scientific Masterclass of the University of Groningen, the Netherlands, respectively. The Lifelines NEXT cohort study received funds from the University Medical Center Groningen Hereditary Metabolic Diseases Fund, Health~Holland (Top Sector Life Sciences and Health), the Ubbo Emmius Foundation, the European Union, the Northern Netherlands Alliance (SNN), the provinces of Friesland and Groningen, the municipality of Groningen, Philips, and the Société des Produits Nestlé.
- De-Cock Hadders foundation (2021-57) International Society of Research in Human Milk and Lactation (ISRHML personal grant)
- Dutch Research Council (NWO gravitation grant Exposome-NL 024.004.017)
- De-Cock Hadders foundation (2021-08) University of Groningen Graduate School of Medical Sciences (scholarship)
- EASI-Genomics (grant PID7780) Winston Bakker Fonds (WB-08) University of Groningen Junior Scientific Masterclass (scholarship)
- Dutch Research Council (NWO-VICI grant VI.C.202.022) Dutch Research Council (NWO gravitation grant Netherlands Organ-on-Chip Initiative 024.003.001) European Research Council (ERC consolidator grant 101001678)
Collapse
Affiliation(s)
- Johanne E Spreckels
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Asier Fernández-Pato
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Marloes Kruk
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Alexander Kurilshikov
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Sanzhima Garmaeva
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Trishla Sinha
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Hiren Ghosh
- Medical Center - University of Freiburg, Institute for Infection Prevention and Hospital Epidemiology, Freiburg, Germany
| | - Hermie Harmsen
- Department of Medical Microbiology, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Jingyuan Fu
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
- Department of Pediatrics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Ranko Gacesa
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
9
|
Andres SF, Zhang Y, Kuhn M, Scottoline B. Building better barriers: how nutrition and undernutrition impact pediatric intestinal health. Front Immunol 2023; 14:1192936. [PMID: 37545496 PMCID: PMC10401430 DOI: 10.3389/fimmu.2023.1192936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/26/2023] [Indexed: 08/08/2023] Open
Abstract
Chronic undernutrition is a major cause of death for children under five, leaving survivors at risk for adverse long-term consequences. This review focuses on the role of nutrients in normal intestinal development and function, from the intestinal epithelium, to the closely-associated mucosal immune system and intestinal microbiota. We examine what is known about the impacts of undernutrition on intestinal physiology, with focus again on the same systems. We provide a discussion of existing animal models of undernutrition, and review the evidence demonstrating that correcting undernutrition alone does not fully ameliorate effects on intestinal function, the microbiome, or growth. We review efforts to treat undernutrition that incorporate data indicating that improved recovery is possible with interventions focused not only on delivery of sufficient energy, macronutrients, and micronutrients, but also on efforts to correct the abnormal intestinal microbiome that is a consequence of undernutrition. Understanding of the role of the intestinal microbiome in the undernourished state and correction of the phenotype is both complex and a subject that holds great potential to improve recovery. We conclude with critical unanswered questions in the field, including the need for greater mechanistic research, improved models for the impacts of undernourishment, and new interventions that incorporate recent research gains. This review highlights the importance of understanding the mechanistic effects of undernutrition on the intestinal ecosystem to better treat and improve long-term outcomes for survivors.
Collapse
Affiliation(s)
- Sarah F. Andres
- Division of Pediatric Gastroenterology, Department of Pediatrics, Oregon Health and Science University, Portland, OR, United States
| | - Yang Zhang
- Division of Pediatric Gastroenterology, Department of Pediatrics, Oregon Health and Science University, Portland, OR, United States
| | - Madeline Kuhn
- Division of Pediatric Gastroenterology, Department of Pediatrics, Oregon Health and Science University, Portland, OR, United States
| | - Brian Scottoline
- Division of Neonatology, Department of Pediatrics, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
10
|
Reniker LN, Frazer LC, Good M. Key biologically active components of breast milk and their beneficial effects. Semin Pediatr Surg 2023; 32:151306. [PMID: 37276783 PMCID: PMC10330649 DOI: 10.1016/j.sempedsurg.2023.151306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Maternal breast milk is the penultimate nutritional source for term and preterm neonates. Its composition is highly complex and includes multiple factors that enhance the development of nearly every neonatal organ system leading to both short- and long-term health benefits. Intensive research is focused on identifying breast milk components that enhance infant health. However, this research is complicated by the significant impact of maternal factors and the processing of pumped breast milk on bioactive ingredients. Optimizing enteral nutrition is particularly important for preterm neonates who miss the transplacental acquisition of nutrients in the third trimester of pregnancy and are at risk for illnesses associated with gut barrier dysfunction, including sepsis and necrotizing enterocolitis. In this review, we will discuss the health benefits of breast milk and its bioactive components.
Collapse
Affiliation(s)
- Laura N Reniker
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA, 27599
| | - Lauren C Frazer
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA, 27599
| | - Misty Good
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA, 27599.
| |
Collapse
|
11
|
Samarra A, Esteban-Torres M, Cabrera-Rubio R, Bernabeu M, Arboleya S, Gueimonde M, Collado MC. Maternal-infant antibiotic resistance genes transference: what do we know? Gut Microbes 2023; 15:2194797. [PMID: 37020319 PMCID: PMC10078139 DOI: 10.1080/19490976.2023.2194797] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 03/21/2023] [Indexed: 04/07/2023] Open
Abstract
Resistance to antibiotics is becoming a worldwide threat as infections caused by multidrug-resistant pathogenic microorganisms can overcome antibiotic treatments and spread quickly in the population. In the context of early life, newborns are at increased risk as their immune system is still under development, so infections and acquisition of resistance during childhood have short- and long-term consequences for the health. The moment of birth is the first exposure of infants to possible antibiotic-resistant microorganisms that may colonize their gut and other body sites. Different factors including mode of delivery, previous antibiotic exposure of the mother, gestational age and consumption of antibiotics in early-life have been described to modulate the neonate's microbiota, and thus, the resistome. Other factors, such as lactation, also impact the establishment and development of gut microbiota, but little is known about the role of breastmilk in transferring Antibiotic Resistant Genes (ARG). A deeper understanding of vertical transmission of antibiotic resistance from mothers to their offspring is necessary to determine the most effective strategies for reducing antibiotic resistance in the early life. In this review, we aim to present the current perspective on antibiotic resistances in mother-infant dyads, as well as a new insight on the study of the human gut and breastmilk resistome, and current strategies to overcome this public health problem, toward highlighting the gaps of knowledge that still need to be closed.
Collapse
Affiliation(s)
- Anna Samarra
- Department of Biotechnology, Institute of Agrochemistry and Food Technology- National Research Council (IATA-CSIC), Valencia, Spain
| | - Maria Esteban-Torres
- Department of Biotechnology, Institute of Agrochemistry and Food Technology- National Research Council (IATA-CSIC), Valencia, Spain
| | - Raul Cabrera-Rubio
- Department of Biotechnology, Institute of Agrochemistry and Food Technology- National Research Council (IATA-CSIC), Valencia, Spain
| | - Manuel Bernabeu
- Department of Biotechnology, Institute of Agrochemistry and Food Technology- National Research Council (IATA-CSIC), Valencia, Spain
- Vicerectorat de Recerca, Universitat de Barcelona (UB), Barcelona, Spain
| | - Silvia Arboleya
- Department of Microbiology and Biochemistry, Dairy Research Institute- National Research Council (IPLA-CSIC), Villaviciosa, Spain
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry, Dairy Research Institute- National Research Council (IPLA-CSIC), Villaviciosa, Spain
| | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology- National Research Council (IATA-CSIC), Valencia, Spain
| |
Collapse
|
12
|
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
|
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: 2.7] [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
|
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: 1.0] [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
|
16
|
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
|
17
|
Exploring the Potential of Human Milk and Formula Milk on Infants’ Gut and Health. Nutrients 2022; 14:nu14173554. [PMID: 36079814 PMCID: PMC9460722 DOI: 10.3390/nu14173554] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 11/21/2022] Open
Abstract
Early-life gut microbiota plays a role in determining the health and risk of developing diseases in later life. Various perinatal factors have been shown to contribute to the development and establishment of infant gut microbiota. One of the important factors influencing the infant gut microbial colonization and composition is the mode of infant feeding. While infant formula milk has been designed to resemble human milk as much as possible, the gut microbiome of infants who receive formula milk differs from that of infants who are fed human milk. A diverse microbial population in human milk and the microbes seed the infant gut microbiome. Human milk contains nutritional components that promote infant growth and bioactive components, such as human milk oligosaccharides, lactoferrin, and immunoglobulins, which contribute to immunological development. In an attempt to encourage the formation of a healthy gut microbiome comparable to that of a breastfed infant, manufacturers often supplement infant formula with prebiotics or probiotics, which are known to have a bifidogenic effect and can modulate the immune system. This review aims to elucidate the roles of human milk and formula milk on infants’ gut and health.
Collapse
|
18
|
Firmicutes, Bacteroidetes and Actinobacteria in Human Milk and Maternal Adiposity. Nutrients 2022; 14:nu14142887. [PMID: 35889844 PMCID: PMC9315738 DOI: 10.3390/nu14142887] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/02/2022] [Accepted: 07/12/2022] [Indexed: 12/13/2022] Open
Abstract
The main objective was to explore the relationship between the microbiota of human milk and adiposity in Mexican mothers during the first lactation stage. Methods: Seventy lactating women were included. Adiposity by anthropometric measurements and by bioelectric impedance was obtained. The donation of human milk was requested, from which bacterial DNA was extracted and qPCR of the 16S region was performed. The Mann–Whitney U test, Spearman and Pearson correlations, and multiple linear regressions models were also calculated. Results: The median percentage of Bacteroidetes had a direct and significant correlation with normal adiposity, current BMI, waist circumference, and body fat percentage. The correlation with current BMI became significantly inverse in women with BMI ≥ 25. In women with normal BMI, the percentage of Actinobacteria showed a direct and significant correlation with current BMI, waist circumference, and percentage of body fat. Multiple linear regressions showed that pre-pregnancy BMI was the variable with the highest predictive value with the Bacteroidetes phyla in normal BMI and in BMI ≥ 25. Conclusions: the adiposity of the woman before pregnancy and during lactation would have an important effect on the abundance of Bacteroidetes and Actinobacteria in human milk.
Collapse
|
19
|
Wong E, Lui K, Day AS, Leach ST. Manipulating the neonatal gut microbiome: current understanding and future perspectives. Arch Dis Child Fetal Neonatal Ed 2022; 107:346-350. [PMID: 34433586 PMCID: PMC9209688 DOI: 10.1136/archdischild-2021-321922] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 08/10/2021] [Indexed: 11/16/2022]
Abstract
The development of a healthy intestinal microbiome following birth contributes to the overall health of the infant during childhood and into adulthood. However, modern birth practices such as caesarean delivery, feeding, antibiotic exposure as well as maternal factors have the potential to greatly impact infant microbiome development. Aberrant microbiome development may be a key factor in the increasing incidence of inflammatory and gut diseases. This review will summarise the current understanding of how modern birth practices may contribute to deficiencies in neonatal gut microbiome development and will also present potential methods of microbiome engineering that aim to ensure the development of a healthy and robust microbiome to protect the host from disease throughout their life.
Collapse
Affiliation(s)
- Emma Wong
- School of Women's and Children's Health, UNSW Medicine, Sydney, NSW, Australia
| | - Kei Lui
- School of Women's and Children's Health, UNSW Medicine, Sydney, NSW, Australia
| | - Andrew S Day
- School of Women's and Children's Health, UNSW Medicine, Sydney, NSW, Australia
- Department of Paediatrics, University of Otago Christchurch, Christchurch, New Zealand
| | - Steven T Leach
- School of Women's and Children's Health, UNSW Medicine, Sydney, NSW, Australia
| |
Collapse
|
20
|
Boudar Z, Sehli S, El Janahi S, Al Idrissi N, Hamdi S, Dini N, Brim H, Amzazi S, Nejjari C, Lloyd-Puryear M, Ghazal H. Metagenomics Approaches to Investigate the Neonatal Gut Microbiome. Front Pediatr 2022; 10:886627. [PMID: 35799697 PMCID: PMC9253679 DOI: 10.3389/fped.2022.886627] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/23/2022] [Indexed: 12/03/2022] Open
Abstract
Early infancy is critical for the development of an infant's gut flora. Many factors can influence microbiota development during the pre- and postnatal periods, including maternal factors, antibiotic exposure, mode of delivery, dietary patterns, and feeding type. Therefore, investigating the connection between these variables and host and microbiome interactions in neonatal development would be of great interest. As the "unculturable" era of microbiome research gives way to an intrinsically multidisciplinary field, microbiome research has reaped the advantages of technological advancements in next-generation sequencing, particularly 16S rRNA gene amplicon and shotgun sequencing, which have considerably expanded our knowledge about gut microbiota development during early life. Using omics approaches to explore the neonatal microbiome may help to better understand the link between the microbiome and newborn diseases. Herein, we summarized the metagenomics methods and tools used to advance knowledge on the neonatal microbiome origin and evolution and how the microbiome shapes early and late individuals' lives for health and disease. The way to overcome limitations in neonatal microbiome studies will be discussed.
Collapse
Affiliation(s)
- Zakia Boudar
- Department of Fundamental Sciences, School of Medicine, Mohammed VI University of Health Sciences, Casablanca, Morocco
| | - Sofia Sehli
- Department of Fundamental Sciences, School of Medicine, Mohammed VI University of Health Sciences, Casablanca, Morocco
| | - Sara El Janahi
- Department of Fundamental Sciences, School of Medicine, Mohammed VI University of Health Sciences, Casablanca, Morocco
| | - Najib Al Idrissi
- Department of Surgery, Faculty of Medicine, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - Salsabil Hamdi
- Laboratory of Genomics and Bioinformatics, School of Pharmacy, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - Nouzha Dini
- Mother and Child Department, Cheikh Khalifa International University Hospital, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - Hassan Brim
- Department of Pathology, Howard University, Washington, DC, United States
| | - Saaïd Amzazi
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, and Genomic Center of Human Pathologies, Faculty of Medicine and Pharmacy, Mohammed V University, Rabat, Morocco
| | - Chakib Nejjari
- Department of Epidemiology and Biostatistics, International School of Public Health, Mohammed VI University of Health Sciences, Casablanca, Morocco
- Department of Epidemiology and Public Health, Faculty of Medicine, University Sidi Mohammed Ben Abdellah, Fez, Morocco
| | | | - Hassan Ghazal
- Department of Fundamental Sciences, School of Medicine, Mohammed VI University of Health Sciences, Casablanca, Morocco
- National Center for Scientific and Technical Research, Rabat, Morocco
| |
Collapse
|
21
|
Samuel TM, Thielecke F, Lavalle L, Chen C, Fogel P, Giuffrida F, Dubascoux S, Martínez-Costa C, Haaland K, Marchini G, Agosti M, Rakza T, Costeira MJ, Picaud JC, Billeaud C, Thakkar SK. Mode of Neonatal Delivery Influences the Nutrient Composition of Human Milk: Results From a Multicenter European Cohort of Lactating Women. Front Nutr 2022; 9:834394. [PMID: 35464009 PMCID: PMC9033294 DOI: 10.3389/fnut.2022.834394] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/18/2022] [Indexed: 12/03/2022] Open
Abstract
Background The effect of the mode of neonatal delivery (cesarean or vaginal) on the nutrient composition of human milk (HM) has rarely been studied. Given the increasing prevalence of cesarean section (C-section) globally, understanding the impact of C-section vs. vaginal delivery on the nutrient composition of HM is fundamental when HM is the preferred source of infant food during the first 4 postnatal months. Objective This study aimed to evaluate the association between mode of delivery and nutrient composition of HM in the first 4 months of life. Design Milk samples were obtained from 317 healthy lactating mothers as part of an exploratory analyses within a multicenter European longitudinal cohort (ATLAS cohort) to study the HM composition, and its potential association with the mode of delivery. We employed traditional mixed models to study individual nutrient associations adjusted for mother’s country, infant birth weight, parity, and gestational age, and complemented it, for the first time, with a multidimensional data analyses approach (non-negative tensor factorization, NTF) to examine holistically how patterns of multiple nutrients and changes over time are associated with the delivery mode. Results Over the first 4 months, nutrient profiles in the milk of mothers who delivered vaginally (n = 237) showed significantly higher levels of palmitoleic acid (16:1n-7), stearic acid (18:0), oleic acid (18:1n-9), arachidic acid (20:0), alpha-linolenic acid (18:3n-3), eicosapentaenoic acid (20:5n-3), docosahexenoic acid (22:6n-3), erucic acid (22:1n-9), monounsaturated fatty acids (MUFA)%, calcium, and phosphorus, whereas the ratios of arachidonic acid/docosahexaenoic acid (ARA/DHA) and n-6/n-3, as well as polyunsaturated fatty acids (PUFA)% were higher in milk from women who had C-sections, in the unadjusted analyses (p < 0.05 for all), but did not retain significance when adjusted for confounders in the mixed models. Using a complementary multidimension data analyses approach (NTF), we show few similar patterns wherein a group of mothers with a high density of C-sections showed increased values for PUFA%, n-6/n-3, and ARA/DHA ratios, but decreased values of MUFA%, 20:1n-9, iodine, and fucosyl-sialyl-lacto-N-tetraose 2 during the first 4 months of lactation. Conclusion Our data provide preliminary insights on differences in concentrations of several HM nutrients (predominantly fatty acids) among women who delivered via C-section. Although these effects tend to disappear after adjustment for confounders, given the similar patterns observed using two different data analytical approaches, these preliminary findings warrant further confirmation and additional insight on the biological and clinical effects related to such differences early in life.
Collapse
Affiliation(s)
- Tinu M. Samuel
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Frank Thielecke
- Department of Health Promotion, Swiss Distance University of Applied Sciences, Regensdorf, Brig, Switzerland
| | - Luca Lavalle
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Cheng Chen
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | | | | | - Stephane Dubascoux
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | | | | | | | | | - Thameur Rakza
- Centre d’Investigation Clinique de Lille, Hôpital Jeanne de Flandre, Lille, France
| | | | | | | | - Sagar K. Thakkar
- Nestlé Research, Société des Produits Nestlé S.A., Singapore, Singapore
- *Correspondence: Sagar K. Thakkar,
| |
Collapse
|
22
|
Lyons KE, Shea CAO, Grimaud G, Ryan CA, Dempsey E, Kelly AL, Ross RP, Stanton C. The human milk microbiome aligns with lactation stage and not birth mode. Sci Rep 2022; 12:5598. [PMID: 35379843 PMCID: PMC8979980 DOI: 10.1038/s41598-022-09009-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 03/03/2022] [Indexed: 12/13/2022] Open
Abstract
We analysed the human milk microbiome in a cohort of 80 lactating women and followed the dynamics in taxa over the course of lactation from birth to 6 months. Two hundred and thirty one milk samples were collected from full-term lactating women at 1, 4, 8 and 24 weeks following birth and analysed for microbiota composition using 16S rRNA sequencing. A significant decrease in milk microbiota diversity was observed throughout the first 6 months of lactation, with the greatest difference seen between week 8 and week 24. Nine genera predominated in milk over lactation from week 1 to week 24, comprising of Staphylococcus, Streptococcus, Pseudomonas, Acinetobacter, Bifidobacterium, Mesorhizobium, Brevundimonas, Flavobacterium, and Rhodococcus; however, fluctuations in these core genera were apparent over time. There was a significant effect of stage of lactation on the microbiome, while no effect of birth mode, infant sex and maternal BMI was observed throughout lactation. Streptococcus had the highest mean relative abundance at week 1 and 24 (17.3% and 24% respectively), whereas Pseudomonas predominated at week 4 (22%) and week 8 (19%). Bifidobacterium and Lactobacillus had the highest mean relative abundance at week 4 (5% and 1.4% respectively), and occurred at a relative abundance of ≤ 1% at all other time points. A decrease in milk microbiota diversity throughout lactation was also observed. This study concluded that lactation stage was the primary driving factor in milk microbiota compositional changes over lactation from birth to 6 months, while mode of delivery was not a factor driving compositional changes throughout human lactation.
Collapse
|
23
|
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: 0.7] [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
|
24
|
Human Milk Oligosaccharides and Bacterial Profile Modulate Infant Body Composition during Exclusive Breastfeeding. Int J Mol Sci 2022; 23:ijms23052865. [PMID: 35270006 PMCID: PMC8911220 DOI: 10.3390/ijms23052865] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/23/2022] [Accepted: 03/02/2022] [Indexed: 02/06/2023] Open
Abstract
Human milk is a complex and variable ecosystem fundamental to the development of newborns. This study aimed to investigate relationships between human milk oligosaccharides (HMO) and human milk bacterial profiles and infant body composition. Human milk samples (n = 60) were collected at two months postpartum. Infant and maternal body composition was measured with bioimpedance spectroscopy. Human milk bacterial profiles were assessed using full-length 16S rRNA gene sequencing and 19 HMOs were quantitated using high-performance liquid chromatography. Relative abundance of human milk bacterial taxa were significantly associated with concentrations of several fucosylated and sialylated HMOs. Individual human milk bacteria and HMO intakes and concentrations were also significantly associated with infant anthropometry, fat-free mass, and adiposity. Furthermore, when data were stratified based on maternal secretor status, some of these relationships differed significantly among infants born to secretor vs non-secretor mothers. In conclusion, in this pilot study the human milk bacterial profile and HMO intakes and concentrations were significantly associated with infant body composition, with associations modified by secretor status. Future research designed to increase the understanding of the mechanisms by which HMO and human milk bacteria modulate infant body composition should include intakes in addition to concentrations.
Collapse
|
25
|
Moya-Alvarez V, Sansonetti PJ. Understanding the pathways leading to gut dysbiosis and enteric environmental dysfunction in infants: the influence of maternal dysbiosis and other microbiota determinants during early life. FEMS Microbiol Rev 2022; 46:6516326. [PMID: 35088084 DOI: 10.1093/femsre/fuac004] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 12/10/2021] [Accepted: 01/25/2022] [Indexed: 11/13/2022] Open
Abstract
Maternal environmental enteric dysfunction (EED) encompasses undernutrition with an inflammatory gut profile, a variable degree of dysbiosis and increased translocation of pathogens in the gut mucosa. Even though recent research findings have shed light on the pathological pathways underlying the establishment of the infant gut dysbiosis, evidence on how maternal EED influences the development of gut dysbiosis and EED in the offspring remains elusive. This review summarizes the current knowledge on the effect of maternal dysbiosis and EED on infant health, and explores recent progress in unraveling the mechanisms of acquisition of a dysbiotic gut microbiota in the offspring. In Western communities, maternal inoculum, delivery mode, perinatal antibiotics, feeding practices, and infections are the major drivers of the infant gut microbiota during the first two years of life. In other latitudes, the infectious burden and maternal malnutrition might introduce further risk factors for infant gut dysbiosis. Novel tools, such as transcriptomics and metabolomics, have become indispensable to analyze the metabolic environment of the infant in utero and post-partum. Human-milk oligosaccharides have essential prebiotic, antimicrobial, and anti-biofilm properties that might offer additional therapeutic opportunities.
Collapse
Affiliation(s)
- Violeta Moya-Alvarez
- Molecular Microbial Pathogenesis - INSERM U1202, Department of Cell Biology and Infection, 28 rue du Dr. Roux, Institut Pasteur, 75015 Paris, France.,Epidemiology of Emergent Diseases Unit, Global Health Department, 25 rue du Dr. Roux, Institut Pasteur, 75015 Paris, France
| | - Philippe J Sansonetti
- Molecular Microbial Pathogenesis - INSERM U1202, Department of Cell Biology and Infection, 28 rue du Dr. Roux, Institut Pasteur, 75015 Paris, France.,Chaire de Microbiologie et Maladies Infectieuses, Collège de France, Paris, France.,The Center for Microbes, Development and Health, Institut Pasteur de Shanghai, China
| |
Collapse
|
26
|
The hidden universe of human milk microbiome: origin, composition, determinants, role, and future perspectives. Eur J Pediatr 2022; 181:1811-1820. [PMID: 35124754 PMCID: PMC9056486 DOI: 10.1007/s00431-022-04383-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 12/14/2022]
Abstract
UNLABELLED Although traditionally considered sterile, human milk is currently recognized as an alive ecosystem that harbors not only bacteria, but also viruses, fungi and yeasts, and minor genera, collectively known as the human milk microbiome (HMM). The seeding of HMM is a complex phenomenon whose dynamics are still a matter of research. Many factors contribute to its determination, both maternal, neonatal, environmental, and related to human milk itself. The transmission of microorganisms to the infant through breastfeeding may impact its present and future health, mainly shaping the GI tract microbiome and immune system. The existence and persistence of HMM as a conserved feature among different species may also have an evolutionary meaning, which will become apparent only in evolutionary times. CONCLUSION The complexities of HMM warrant further research in order to deepen our knowledge on its origin, determinants, and impact on infants' health. The practical and translational implications of research on HMM (e.g., reconstitution of donor human milk through inoculation of infant's own mother milk, modulation of HMM through maternal dietary supplementation) should not be overlooked. WHAT IS KNOWN • Human milk harbors a wide variety of microorganisms, ranging from bacteria to viruses, fungi and yeasts, and minor genera. • Human milk microbiome is shaped over time by many factors: maternal, neonatal, environmental, and related to human milk itself. • The transmission of microorganisms through breastfeeding may impact the infant's present and future health. WHAT IS NEW • We provide an overview on human milk microbiome, hopefully encouraging physicians to consider it among the other better-known breastfeeding benefits. • Further studies, with standardized and rigorous study designs to enhance accuracy and reproducibility of the results, are needed to deepen our knowledge of the human milk microbiota and its role in newborn and infant's health.
Collapse
|
27
|
The Maternal Milk Microbiome in Mammals of Different Types and Its Potential Role in the Neonatal Gut Microbiota Composition. Animals (Basel) 2021; 11:ani11123349. [PMID: 34944125 PMCID: PMC8698027 DOI: 10.3390/ani11123349] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/20/2021] [Accepted: 11/22/2021] [Indexed: 12/19/2022] Open
Abstract
Maternal milk, a main source of nutrition for neonates in early life, has attracted attention. An increasing number of studies have found that maternal milk has a high microbial diversity, as well as factors that might influence this diversity. However, there is a lack of knowledge regarding the effects of host diet and phylogeny on maternal milk microbes and the contribution of the maternal milk microbiota to the neonatal gut microbiota. Here, we analyzed the maternal milk and fecal microbiota of nine species (lion, dog, panda, human, mouse, rhesus macaque, cow, goat, and rabbit) of mammals of three type groups (herbivore, omnivore, and carnivore) using 16S rRNA amplicon sequencing. Our study provided evidence of host diet and phylogeny on the maternal milk microbiota. Moreover, functional prediction revealed that the carnivores had a significantly higher percentage of base excision repair, glycerolipid metabolism, taurine and hypotaurine metabolism, inorganic ion transport and metabolism, and nucleotide metabolism; while arginine and proline metabolism showed enrichment in the herbivore group. Source-tracking analysis showed that the contributions of bacteria from maternal milk to the microbiota of neonates of different mammals were different at day 3 after neonatal birth. Overall, our findings provided a theoretical basis for the maternal milk microbiota to affect neonatal fecal microbiota at day 3 after neonatal birth.
Collapse
|
28
|
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
|
29
|
Vizzari G, Morniroli D, Ceroni F, Verduci E, Consales A, Colombo L, Cerasani J, Mosca F, Giannì ML. Human Milk, More Than Simple Nourishment. CHILDREN (BASEL, SWITZERLAND) 2021; 8:863. [PMID: 34682128 PMCID: PMC8535116 DOI: 10.3390/children8100863] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/18/2021] [Accepted: 09/19/2021] [Indexed: 12/24/2022]
Abstract
Human breast milk not only has nutritional properties but also holds a functional role. It contains various bioactive factors (lactoferrin, lysozyme, leukocytes, immunoglobulins, cytokines, hormones, human milk oligosaccharides, microbiome, microRNAs and stem cells) shown to contribute to several short- and long-term health outcomes. Some of these factors appear to be involved in the infant's neuro-cognitive development, anti-oncogenic processes, cellular communication and differentiation. Furthermore, breast milk is increasingly recognized to have dynamic characteristics and to play a fundamental role in the cross-talking mother-neonate. This narrative review aims to provide a summary and an update on these bioactive substances, exploring their functions mainly on immunomodulation, microbiome and virome development. Although the knowledge about breast milk potentiality has significantly improved, leading to discovering unexpected functions, the exact mechanisms with which breast milk exercises its bioactivity have not been completely clarified. This can represent a fertile ground for exploring and understanding the complexity behind these functional elements to develop new therapeutic strategies.
Collapse
Affiliation(s)
- Giulia Vizzari
- Department of Clinical Sciences and Community Health, University of Milan, Via Commenda 19, 20122 Milan, Italy; (G.V.); (D.M.); (F.C.); (A.C.); (J.C.); (F.M.)
| | - Daniela Morniroli
- Department of Clinical Sciences and Community Health, University of Milan, Via Commenda 19, 20122 Milan, Italy; (G.V.); (D.M.); (F.C.); (A.C.); (J.C.); (F.M.)
| | - Federica Ceroni
- Department of Clinical Sciences and Community Health, University of Milan, Via Commenda 19, 20122 Milan, Italy; (G.V.); (D.M.); (F.C.); (A.C.); (J.C.); (F.M.)
| | - Elvira Verduci
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy;
- Department of Health Sciences, University of Milan, 20154 Milan, Italy
| | - Alessandra Consales
- Department of Clinical Sciences and Community Health, University of Milan, Via Commenda 19, 20122 Milan, Italy; (G.V.); (D.M.); (F.C.); (A.C.); (J.C.); (F.M.)
| | - Lorenzo Colombo
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico NICU, Via Commenda 12, 20122 Milan, Italy;
| | - Jacopo Cerasani
- Department of Clinical Sciences and Community Health, University of Milan, Via Commenda 19, 20122 Milan, Italy; (G.V.); (D.M.); (F.C.); (A.C.); (J.C.); (F.M.)
| | - Fabio Mosca
- Department of Clinical Sciences and Community Health, University of Milan, Via Commenda 19, 20122 Milan, Italy; (G.V.); (D.M.); (F.C.); (A.C.); (J.C.); (F.M.)
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico NICU, Via Commenda 12, 20122 Milan, Italy;
| | - Maria Lorella Giannì
- Department of Clinical Sciences and Community Health, University of Milan, Via Commenda 19, 20122 Milan, Italy; (G.V.); (D.M.); (F.C.); (A.C.); (J.C.); (F.M.)
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico NICU, Via Commenda 12, 20122 Milan, Italy;
| |
Collapse
|
30
|
Sanjulián L, Lamas A, Barreiro R, Cepeda A, Fente CA, Regal P. Bacterial Diversity of Breast Milk in Healthy Spanish Women: Evolution from Birth to Five Years Postpartum. Nutrients 2021; 13:2414. [PMID: 34371924 PMCID: PMC8308733 DOI: 10.3390/nu13072414] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/07/2021] [Accepted: 07/09/2021] [Indexed: 12/27/2022] Open
Abstract
The objective of this work was to characterize the microbiota of breast milk in healthy Spanish mothers and to investigate the effects of lactation time on its diversity. A total of ninety-nine human milk samples were collected from healthy Spanish women and were assessed by means of next-generation sequencing of 16S rRNA amplicons and by qPCR. Firmicutes was the most abundant phylum, followed by Bacteroidetes, Actinobacteria, and Proteobacteria. Accordingly, Streptococcus was the most abundant genus. Lactation time showed a strong influence in milk microbiota, positively correlating with Actinobacteria and Bacteroidetes, while Firmicutes was relatively constant over lactation. 16S rRNA amplicon sequencing showed that the highest alpha-diversity was found in samples of prolonged lactation, along with wider differences between individuals. As for milk nutrients, calcium, magnesium, and selenium levels were potentially associated with Streptococcus and Staphylococcus abundance. Additionally, Proteobacteria was positively correlated with docosahexaenoic acid (DHA) levels in breast milk, and Staphylococcus with conjugated linoleic acid. Conversely, Streptococcus and trans-palmitoleic acid showed a negative association. Other factors such as maternal body mass index or diet also showed an influence on the structure of these microbial communities. Overall, human milk in Spanish mothers appeared to be a complex niche shaped by host factors and by its own nutrients, increasing in diversity over time.
Collapse
Affiliation(s)
| | - Alexandre Lamas
- Department of Analytical Chemistry, Nutrition and Bromatology, Faculty of Veterinary Science, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (L.S.); (R.B.); (A.C.); (C.A.F.)
| | | | | | | | - Patricia Regal
- Department of Analytical Chemistry, Nutrition and Bromatology, Faculty of Veterinary Science, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (L.S.); (R.B.); (A.C.); (C.A.F.)
| |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW To perform a nonsystematic review of the literature on the role of breastfeeding as primary prevention tool for allergic diseases. RECENT FINDINGS Human milk contains vast amounts of biologically active components that have a significant impact on the development of the gut microbiota. Exclusively breastfed infants show a different microbiota, with a predominance of Bifidobacterium species in their intestines.The mechanisms underlying the antiallergic effects of human milk are most probably complex, as human milk contains not only nutritional substances but also functional molecules including polysaccharides, cytokines, proteins, and other components which can produce an epigenetic modulation of the innate and adaptive immune responses of the infant in very early life. SUMMARY Currently, there is not sufficient strong evidence to guarantee its effectiveness in allergy prevention and therefore the main international scientific societies still do not count it among the recognized primary prevention strategies of allergy.
Collapse
Affiliation(s)
- Maurizio Mennini
- Multifactorial and Systemic Diseases Research Area, Predictive and Preventive Medicine Research Unit, Division of Allergy Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | | | | |
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: 0.8] [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
|
Coscia A, Bardanzellu F, Caboni E, Fanos V, Peroni DG. When a Neonate Is Born, So Is a Microbiota. Life (Basel) 2021; 11:life11020148. [PMID: 33669262 PMCID: PMC7920069 DOI: 10.3390/life11020148] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/09/2021] [Accepted: 02/11/2021] [Indexed: 02/06/2023] Open
Abstract
In recent years, the role of human microbiota as a short- and long-term health promoter and modulator has been affirmed and progressively strengthened. In the course of one’s life, each subject is colonized by a great number of bacteria, which constitute its specific and individual microbiota. Human bacterial colonization starts during fetal life, in opposition to the previous paradigm of the “sterile womb”. Placenta, amniotic fluid, cord blood and fetal tissues each have their own specific microbiota, influenced by maternal health and habits and having a decisive influence on pregnancy outcome and offspring outcome. The maternal microbiota, especially that colonizing the genital system, starts to influence the outcome of pregnancy already before conception, modulating fertility and the success rate of fertilization, even in the case of assisted reproduction techniques. During the perinatal period, neonatal microbiota seems influenced by delivery mode, drug administration and many other conditions. Special attention must be reserved for early neonatal nutrition, because breastfeeding allows the transmission of a specific and unique lactobiome able to modulate and positively affect the neonatal gut microbiota. Our narrative review aims to investigate the currently identified pre- and peri-natal factors influencing neonatal microbiota, before conception, during pregnancy, pre- and post-delivery, since the early microbiota influences the whole life of each subject.
Collapse
Affiliation(s)
- Alessandra Coscia
- Neonatology Unit, Department of Public Health and Pediatrics, Università degli Studi di Torino, 10124 Turin, Italy;
| | - Flaminia Bardanzellu
- Neonatal Intensive Care Unit, Department of Surgical Sciences, AOU and University of Cagliari, SS 554 km 4,500, 09042 Monserrato, Italy; (E.C.); (V.F.)
- Correspondence:
| | - Elisa Caboni
- Neonatal Intensive Care Unit, Department of Surgical Sciences, AOU and University of Cagliari, SS 554 km 4,500, 09042 Monserrato, Italy; (E.C.); (V.F.)
| | - Vassilios Fanos
- Neonatal Intensive Care Unit, Department of Surgical Sciences, AOU and University of Cagliari, SS 554 km 4,500, 09042 Monserrato, Italy; (E.C.); (V.F.)
| | - Diego Giampietro Peroni
- Clinical and Experimental Medicine Department, Section of Pediatrics, University of Pisa, Via Roma, 55, 56126 Pisa PI, Italy;
| |
Collapse
|
34
|
Carr LE, Virmani MD, Rosa F, Munblit D, Matazel KS, Elolimy AA, Yeruva L. Role of Human Milk Bioactives on Infants' Gut and Immune Health. Front Immunol 2021; 12:604080. [PMID: 33643310 PMCID: PMC7909314 DOI: 10.3389/fimmu.2021.604080] [Citation(s) in RCA: 157] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 01/22/2021] [Indexed: 12/26/2022] Open
Abstract
Exclusive human milk feeding of the newborn is recommended during the first 6 months of life to promote optimal health outcomes during early life and beyond. Human milk contains a variety of bioactive factors such as hormones, cytokines, leukocytes, immunoglobulins, lactoferrin, lysozyme, stem cells, human milk oligosaccharides (HMOs), microbiota, and microRNAs. Recent findings highlighted the potential importance of adding HMOs into infant formula for their roles in enhancing host defense mechanisms in neonates. Therefore, understanding the roles of human milk bioactive factors on immune function is critical to build the scientific evidence base around breastfeeding recommendations, and to enhance positive health outcomes in formula fed infants through modifications to formulas. However, there are still knowledge gaps concerning the roles of different milk components, the interactions between the different components, and the mechanisms behind health outcomes are poorly understood. This review aims to show the current knowledge about HMOs, milk microbiota, immunoglobulins, lactoferrin, and milk microRNAs (miRNAs) and how these could have similar mechanisms of regulating gut and microbiota function. It will also highlight the knowledge gaps for future research.
Collapse
Affiliation(s)
- Laura E. Carr
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Arkansas Children's Nutrition Center, Little Rock, AR, United States
| | - Misty D. Virmani
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Fernanda Rosa
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Arkansas Children's Nutrition Center, Little Rock, AR, United States
| | - Daniel Munblit
- Department of Pediatrics and Pediatric Infectious Diseases, Institute of Child's Health, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
- Inflammation, Repair and Development Section, Faculty of Medicine, Imperial College London, National Heart and Lung Institute, London, United Kingdom
- Research and Clinical Center for Neuropsychiatry, Moscow, Russia
| | | | - Ahmed A. Elolimy
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Arkansas Children's Nutrition Center, Little Rock, AR, United States
| | - Laxmi Yeruva
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Arkansas Children's Nutrition Center, Little Rock, AR, United States
- Arkansas Children's Research Institute, Little Rock, AR, United States
| |
Collapse
|
35
|
Cortes-Macías E, Selma-Royo M, García-Mantrana I, Calatayud M, González S, Martínez-Costa C, Collado MC. Maternal Diet Shapes the Breast Milk Microbiota Composition and Diversity: Impact of Mode of Delivery and Antibiotic Exposure. J Nutr 2021; 151:330-340. [PMID: 33188413 PMCID: PMC7850106 DOI: 10.1093/jn/nxaa310] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/21/2020] [Accepted: 09/16/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Breast milk is a complex biofluid that provides nutrients and bioactive agents, including bacteria, for the development of the infant gut microbiota. However, the impact of maternal diet and other factors, such as mode of delivery and antibiotic exposure, on the breast milk microbiota has yet to be understood. OBJECTIVES This study aimed to examine the association between maternal diet and breast milk microbiota and to ascertain the potential role of mode of delivery and antibiotic exposure. METHODS In a cross-sectional study of the MAMI cohort, breast milk microbiota profiling was assessed in 120 samples from healthy mothers by 16S rRNA gene sequencing. Maternal dietary information was recorded through an FFQ, and clinical characteristics, including mode of delivery, antibiotic exposure, and exclusive breastfeeding, were collected. RESULTS Maternal diet was grouped into 2 clusters: Cluster I (high intake of plant protein, fiber, and carbohydrates), and Cluster II (high intake of animal protein and lipids). Breast milk microbiota was shaped by maternal dietary clusters. Staphylococcus and Bifidobacterium were associated with carbohydrate intake whereas the Streptococcus genus was associated with intakes of the n-3 PUFAs [EPA and docosapentaenoic acid (22:5ω-3)]. Mode of delivery and antibiotic exposure influenced breast milk microbiota in a diet cluster-dependent manner. Differences between/among the maternal dietary clusters were found in the milk microbiota of the cesarean-section (C-section)/antibiotic group, whereas no differences were observed in vaginal births. Lower abundances of Lactobacillus, Bacteroides, and Sediminibacterium genera were observed in Cluster II/C-section/antibiotic exposure compared with the other groups. CONCLUSIONS Maternal diet shapes the composition and diversity of breast milk microbiota, with the most important contributions coming from dietary fiber and both plant and animal protein intakes. The relation between the maternal diet and the milk microbiota needs further research because it has a key impact on infant microbiota development and contributes to infant health outcomes in the short and long term.This trial was registered at clinicaltrials.gov as NCT03552939.
Collapse
Affiliation(s)
- Erika Cortes-Macías
- Institute of Agrochemistry and Food Technology (IATA-CSIC), National Research Council, Valencia, Spain
| | - Marta Selma-Royo
- Institute of Agrochemistry and Food Technology (IATA-CSIC), National Research Council, Valencia, Spain
| | - Izaskun García-Mantrana
- Institute of Agrochemistry and Food Technology (IATA-CSIC), National Research Council, Valencia, Spain
| | - Marta Calatayud
- Institute of Agrochemistry and Food Technology (IATA-CSIC), National Research Council, Valencia, Spain
| | - Sonia González
- Department of Functional Biology, Faculty of Medicine, University of Oviedo, Oviedo, Spain
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (DIMISA, ISPA), Oviedo, Spain
| | - Cecilia Martínez-Costa
- Department of Pediatrics, School of Medicine, University of Valencia, Valencia, Spain
- Pediatric Gastroenterology and Nutrition Section, Hospital Clínico Universitario Valencia, INCLIVA, Valencia, Spain
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology (IATA-CSIC), National Research Council, Valencia, Spain
| |
Collapse
|
36
|
Ruiz L, Alba C, García-Carral C, Jiménez EA, Lackey KA, McGuire MK, Meehan CL, Foster J, Sellen DW, Kamau-Mbuthia EW, Kamundia EW, Mbugua S, Moore SE, Prentice AM, Gindola K D, Otoo GE, Pareja RG, Bode L, McGuire MA, Williams JE, Rodríguez JM. Comparison of Two Approaches for the Metataxonomic Analysis of the Human Milk Microbiome. Front Cell Infect Microbiol 2021; 11:622550. [PMID: 33842385 PMCID: PMC8027255 DOI: 10.3389/fcimb.2021.622550] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 03/05/2021] [Indexed: 01/04/2023] Open
Abstract
Recent work has demonstrated the existence of large inter-individual and inter-population variability in the microbiota of human milk from healthy women living across variable geographical and socio-cultural settings. However, no studies have evaluated the impact that variable sequencing approaches targeting different 16S rRNA variable regions may have on the human milk microbiota profiling results. This hampers our ability to make meaningful comparisons across studies. In this context, the main purpose of the present study was to re-process and re-sequence the microbiome in a large set of human milk samples (n = 412) collected from healthy women living at diverse international sites (Spain, Sweden, Peru, United States, Ethiopia, Gambia, Ghana and Kenya), by targeting a different 16S rRNA variable region and reaching a larger sequencing depth. Despite some differences between the results obtained from both sequencing approaches were notable (especially regarding alpha and beta diversities and Proteobacteria representation), results indicate that both sequencing approaches revealed a relatively consistent microbiota configurations in the studied cohorts. Our data expand upon the milk microbiota results we previously reported from the INSPIRE cohort and provide, for the first time across globally diverse populations, evidence of the impact that different DNA processing and sequencing approaches have on the microbiota profiles obtained for human milk samples. Overall, our results corroborate some similarities regarding the microbial communities previously reported for the INSPIRE cohort, but some differences were also detected. Understanding the impact of different sequencing approaches on human milk microbiota profiles is essential to enable meaningful comparisons across studies. Clinical Trial Registration www.clinicaltrials.gov, identifier NCT02670278.
Collapse
Affiliation(s)
- Lorena Ruiz
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
- *Correspondence: Lorena Ruiz, ; Juan Miguel Rodriguez,
| | - Claudio Alba
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| | - Cristina García-Carral
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| | - Esther A. Jiménez
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| | - Kimberly A. Lackey
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID, United States
| | - Michelle K. McGuire
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID, United States
| | - Courtney L. Meehan
- Department of Anthropology, Washington State University, Pullman, WA, United States
| | - James Foster
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID, United States
| | - Daniel W. Sellen
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | | | | | - Samwel Mbugua
- Department of Human Nutrition, Egerton University, Nakuru, Kenya
| | - Sophie E. Moore
- Division of Women’s Health, King’s College London, London, United Kingdom
- MRC Unit, Serekunda, Gambia
| | - Andrew M. Prentice
- MRC International Nutrition Group, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Debela Gindola K
- Department of Anthropology, Hawassa University, Hawassa, Ethiopia
| | - Gloria E. Otoo
- Department of Nutrition and Food Science, University of Ghana, Accra, Ghana
| | | | - Lars Bode
- Department of Pediatrics and Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (MOMI CoRE), University of California, San Diego, La Jolla, CA, United States
| | - Mark A. McGuire
- Department of Animal and Veterinary Science, University of Idaho, Moscow, ID, United States
| | - Janet E. Williams
- Department of Animal and Veterinary Science, University of Idaho, Moscow, ID, United States
| | - Juan M. Rodríguez
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
- *Correspondence: Lorena Ruiz, ; Juan Miguel Rodriguez,
| |
Collapse
|
37
|
Egan M, Dempsey E, Ryan CA, Ross RP, Stanton C. The Sporobiota of the Human Gut. Gut Microbes 2021; 13:1-17. [PMID: 33406976 PMCID: PMC7801112 DOI: 10.1080/19490976.2020.1863134] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 12/01/2020] [Accepted: 12/04/2020] [Indexed: 02/04/2023] Open
Abstract
The human gut microbiome is a diverse and complex ecosystem that plays a critical role in health and disease. The composition of the gut microbiome has been well studied across all stages of life. In recent years, studies have investigated the production of endospores by specific members of the gut microbiome. An endospore is a tough, dormant structure formed by members of the Firmicutes phylum, which allows for greater resistance to otherwise inhospitable conditions. This innate resistance has consequences for human health and disease, as well as in biotechnology. In particular, the formation of endospores is strongly linked to antibiotic resistance and the spread of antibiotic resistance genes, also known as the resistome. The term sporobiota has been used to define the spore-forming cohort of a microbial community. In this review, we present an overview of the current knowledge of the sporobiota in the human gut. We discuss the development of the sporobiota in the infant gut and the perinatal factors that may have an effect on vertical transmission from mother to infant. Finally, we examine the sporobiota of critically important food sources for the developing infant, breast milk and powdered infant formula.
Collapse
Affiliation(s)
- Muireann Egan
- Food Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Eugene Dempsey
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Neonatology, Cork University Maternity Hospital, Cork, Ireland
| | - C. Anthony Ryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Neonatology, Cork University Maternity Hospital, Cork, Ireland
| | - R. Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Catherine Stanton
- Food Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
38
|
Łubiech K, Twarużek M. Lactobacillus Bacteria in Breast Milk. Nutrients 2020; 12:E3783. [PMID: 33321792 PMCID: PMC7764098 DOI: 10.3390/nu12123783] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/03/2020] [Accepted: 12/08/2020] [Indexed: 02/07/2023] Open
Abstract
Breast milk is an optimal food for infants and toddlers. The composition of breast milk adapts to the needs of the developing organism, satisfying nutritional needs at an early stage of growth and development. The results of research to date have shown that breast milk is the best food for a child, containing not only nutrients but also biologically active substances that aid in the optimal, proper growth and development of infants. Among the many components of breast milk, an important element is the probiotic microflora, including bacteria of the genus Lactobacillus spp. These organisms exert a multidirectional, health-promoting effect on the body of children who consume breast milk. The number of lactic acid bacteria, including Lactobacillus, colonizing the breast milk environment and their species diversity varies and depends on many factors, both maternal and environmental. Breast milk, as a recommended food for infants, is an important source of probiotic microflora. The aim of this study was to present the current understanding of probiotic bacteria of the genus Lactobacillus present in breast milk.
Collapse
Affiliation(s)
- Katarzyna Łubiech
- Department of Physiology and Toxicology, Faculty of Biological Sciences, Kazimierz Wielki University, Chodkiewicza 30 St., 85-064 Bydgoszcz, Poland;
| | | |
Collapse
|
39
|
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: 3.6] [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
|
40
|
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: 75] [Impact Index Per Article: 15.0] [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
|
41
|
Boix-Amorós A, Hernández-Aguilar MT, Artacho A, Collado MC, Mira A. Human milk microbiota in sub-acute lactational mastitis induces inflammation and undergoes changes in composition, diversity and load. Sci Rep 2020; 10:18521. [PMID: 33116172 PMCID: PMC7595153 DOI: 10.1038/s41598-020-74719-0] [Citation(s) in RCA: 20] [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: 03/02/2020] [Accepted: 10/06/2020] [Indexed: 01/01/2023] Open
Abstract
Sub-acute mastitis (SAM) is a prevalent disease among lactating women, being one of the main reasons for early weaning. Although the etiology and diagnosis of acute mastitis (AM) is well established, little is known about the underlying mechanisms causing SAM. We collected human milk samples from healthy and SAM-suffering mothers, during the course of mastitis and after symptoms disappeared. Total (DNA-based) and active (RNA-based) microbiota were analysed by 16S rRNA gene sequencing and qPCR. Furthermore, mammary epithelial cell lines were exposed to milk pellets, and levels of the pro-inflammatory interleukin IL8 were measured. Bacterial load was significantly higher in the mastitis samples and decreased after clinical symptoms disappeared. Bacterial diversity was lower in SAM milk samples, and differences in bacterial composition and activity were also found. Contrary to AM, the same bacterial species were found in samples from healthy and SAM mothers, although at different proportions, indicating a dysbiotic ecological shift. Finally, mammary epithelial cell exposure to SAM milk pellets showed an over-production of IL8. Our work therefore supports that SAM has a bacterial origin, with increased bacterial loads, reduced diversity and altered composition, which partly recovered after treatment, suggesting a polymicrobial and variable etiology.
Collapse
Affiliation(s)
- Alba Boix-Amorós
- Department of Biotechnology, Spanish National Research Council (IATA-CSIC), Institute of Agrochemistry and Food Technology, Paterna, Spain
- Department of Health and Genomics, Center for Advanced Research in Public Health, FISABIO Foundation, Valencia, Spain
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
| | | | - Alejandro Artacho
- Department of Health and Genomics, Center for Advanced Research in Public Health, FISABIO Foundation, Valencia, Spain
| | - Maria Carmen Collado
- Department of Biotechnology, Spanish National Research Council (IATA-CSIC), Institute of Agrochemistry and Food Technology, Paterna, Spain
| | - Alex Mira
- Department of Biotechnology, Spanish National Research Council (IATA-CSIC), Institute of Agrochemistry and Food Technology, Paterna, Spain.
| |
Collapse
|
42
|
Samuel TM, Zhou Q, Giuffrida F, Munblit D, Verhasselt V, Thakkar SK. Nutritional and Non-nutritional Composition of Human Milk Is Modulated by Maternal, Infant, and Methodological Factors. Front Nutr 2020; 7:576133. [PMID: 33117843 PMCID: PMC7557356 DOI: 10.3389/fnut.2020.576133] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 08/19/2020] [Indexed: 02/06/2023] Open
Abstract
Human milk (HM) is dynamic and shows a high inter- and intra-individual variability. To characterize HM with precision, it is necessary to understand the factors that modulate its composition. The objective of this narrative review is to summarize the maternal, infant and methodological factors that affect HM composition. We searched SCOPUS and PubMed databases for articles related to factors that are known to or could potentially influence HM composition and volume across lactation periods. Our comprehensive review encompasses various maternal-, infant-related, and methodological factors that modulate aspects of HM composition including macro- and micronutrients, vitamins and minerals, as well as volume. The most profound changes were observed in HM lipids and lipophiles. Evidence exists for many of the infant-related factors known to affect the nutritive and non-nutritive components of HM (e.g., birth weight, gestational age, infant age/stage of lactation). In contrast, less is known with respect to maternal factors; where there is either limited research or conflicting evidence (e.g., maternal lifestyle, obstetric history, medical conditions), except for the mother's diet, for which there is a relatively well-established understanding. Equally, although many of the methodological factors (e.g., HM sampling, handling and analytics) are known to impact HM composition, few studies have investigated this as a primary outcome, making it an important area of future research in HM. Here we propose a systematic capture of numerous maternal- and infant-related characteristics to facilitate associative comparisons of HM data within and across studies. Additionally, it would be prudent to standardize the methodological aspects known to affect HM composition in analytics, not only for HM lipids and lipophiles, but also for those nutrients whose variability is yet less well-understood. Defining the factors determining HM composition with accuracy will open perspectives for maternal intervention to optimize milk composition for specific needs of infants.
Collapse
Affiliation(s)
| | | | | | - Daniel Munblit
- Department of Paediatrics and Paediatric Infectious Diseases, Institute of Child's Health, Sechenov First Moscow State Medical University, Moscow, Russia.,Inflammation, Repair and Development Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | | | | |
Collapse
|
43
|
LeMay-Nedjelski L, Asbury MR, Butcher J, Ley SH, Hanley AJ, Kiss A, Unger S, Copeland JK, Wang PW, Stintzi A, O'Connor DL. Maternal Diet and Infant Feeding Practices Are Associated with Variation in the Human Milk Microbiota at 3 Months Postpartum in a Cohort of Women with High Rates of Gestational Glucose Intolerance. J Nutr 2020; 151:320-329. [PMID: 32886107 PMCID: PMC7850034 DOI: 10.1093/jn/nxaa248] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 05/11/2020] [Accepted: 07/27/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Human milk contains a diverse community of bacteria believed to play a role in breast health and inoculation of the infant's gastrointestinal tract. The role of maternal nutrition and infant feeding practices on the human milk microbiota remains poorly understood. OBJECTIVE Our aim was to explore the associations between maternal diet (delivery to 3 mo postpartum), infant feeding practices, and the microbial composition and predicted function in milk from women with varied metabolic status. METHODS This was an exploratory analysis of a previously completed prospective cohort study of women with varying degrees of gestational glucose intolerance (NCT01405547). Milk samples (n = 93 mothers) were collected at 3 mo postpartum. Maternal dietary information (validated food-frequency questionnaire) and infant feeding practices (human milk exclusivity, frequency of direct breastfeeding per day) were collected. V4-16S ribosomal RNA gene sequencing (Illumina MiSeq) was conducted to determine microbiota composition. RESULTS Intake of polyunsaturated fat [β estimate (SE): 0.036 (0.018), P = 0.047] and fiber from grains [0.027 (0.013), P = 0.048] were positively associated with ɑ-diversity (Shannon index) of human milk. Overall microbial composition of human milk clustered based on human milk exclusivity (weighted UniFrac R2 = 0.034, P = 0.015; Bray-Curtis R2 = 0.041, P = 0.007), frequency of direct breastfeeding per day (Bray-Curtis R2 = 0.057, P = 0.026), and maternal fiber intake from grains (Bray-Curtis R2 = 0.055, P = 0.040). Total fiber, fiber from grains, dietary fat, and infant feeding practices were also associated with a number of differentially abundant taxa. The overall composition of predicted microbial functions was associated with total fiber consumption (Bray-Curtis R2 = 0.067, P = 0.036) and human milk exclusivity (Bray-Curtis R2 = 0.041, P = 0.013). CONCLUSIONS Maternal consumption of fiber and fat, as well as mother's infant feeding practices, are important determinants of the human milk microbiota. Understanding whether these microbial changes impact an infant's overall health and development requires future study.
Collapse
Affiliation(s)
- Lauren LeMay-Nedjelski
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada,Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Michelle R Asbury
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada,Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - James Butcher
- Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Sylvia H Ley
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Anthony J Hanley
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Alex Kiss
- Department of Research Design and Biostatistics, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Sharon Unger
- Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada,Department of Pediatrics, Mount Sinai Hospital, Toronto, Ontario, Canada,Division of Neonatology, The Hospital for Sick Children, Toronto, Ontario, Canada,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Julia K Copeland
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, Ontario, Canada
| | - Pauline W Wang
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, Ontario, Canada
| | - Alain Stintzi
- Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | | |
Collapse
|
44
|
Asbury MR, Butcher J, Copeland JK, Unger S, Bando N, Comelli EM, Forte V, Kiss A, LeMay-Nedjelski L, Sherman PM, Stintzi A, Tomlinson C, Wang PW, O'Connor DL. Mothers of Preterm Infants Have Individualized Breast Milk Microbiota that Changes Temporally Based on Maternal Characteristics. Cell Host Microbe 2020; 28:669-682.e4. [PMID: 32888417 DOI: 10.1016/j.chom.2020.08.001] [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: 04/16/2020] [Revised: 07/01/2020] [Accepted: 07/31/2020] [Indexed: 02/08/2023]
Abstract
Mother's milk contains complex microbial communities thought to be important for colonizing a preterm infant's gastrointestinal tract. However, little is known about the microbiota in the preterm mother's milk and factors influencing its composition. We characterized the temporal dynamics of microbial communities in 490 breast milk samples from 86 mothers of preterm infants (born <1,250g) over the first 8 weeks postpartum. Highly individualized microbial communities were identified in each mother's milk that changed temporally with notable alterations in predicted microbial functions. However, pre-pregnancy BMI, delivery mode, and antibiotics were associated with changes in these microbial dynamics. Individual classes of antibiotics and their duration of exposure during prenatal and postpartum periods showed unique relationships with microbial taxa abundance and diversity in mother's milk. These results highlight the temporal complexity of the preterm mother's milk microbiota and its relationship with maternal characteristics as well as the importance of discussing antibiotic stewardship for mothers.
Collapse
Affiliation(s)
- Michelle R Asbury
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Translational Medicine Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - James Butcher
- Department of Biochemistry, Microbiology and Immunology and Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Julia K Copeland
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, ON M5S 3B2, Canada
| | - Sharon Unger
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Rogers Hixon Ontario Human Milk Bank and the Department of Pediatrics, Sinai Health, Toronto, ON M5G 1X5, Canada; Division of Neonatology, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Nicole Bando
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Elena M Comelli
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Joannah and Brian Lawson Centre for Child Nutrition, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Victoria Forte
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Translational Medicine Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Alex Kiss
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON M5T 3M6, Canada; Evaluative and Clinical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - Lauren LeMay-Nedjelski
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Translational Medicine Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Philip M Sherman
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Gastroenterology, Hepatology, and Nutrition, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Alain Stintzi
- Department of Biochemistry, Microbiology and Immunology and Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Christopher Tomlinson
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Translational Medicine Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Division of Neonatology, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Pauline W Wang
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, ON M5S 3B2, Canada
| | - Deborah L O'Connor
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Translational Medicine Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Rogers Hixon Ontario Human Milk Bank and the Department of Pediatrics, Sinai Health, Toronto, ON M5G 1X5, Canada.
| |
Collapse
|
45
|
Quin C, Gibson DL. Human behavior, not race or geography, is the strongest predictor of microbial succession in the gut bacteriome of infants. Gut Microbes 2020; 11:1143-1171. [PMID: 32249675 PMCID: PMC7524360 DOI: 10.1080/19490976.2020.1736973] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Colonization of the gastrointestinal tract with microorganisms during infancy represents a critical control point for shaping life-long immune-mediated disease susceptibility. Abnormal colonization or an imbalance of microbes, termed dysbiosis, is implicated in several diseases. Consequently, recent research has aimed at understanding ways to manipulate a dysbiotic microbiome during infancy to resemble a normal, healthy microbiome. However, one of the fundamental issues in microbiome research is characterizing what a "normal" infant microbiome is based on geography, ethnicity and cultural variations. This review provides a comprehensive account of what is currently known about the infant microbiome from a global context. In general, this review shows that the influence of cultural variations in feeding practices, delivery modes and hygiene are the biggest contributors to microbial variability. Despite geography or race, all humans have similar microbial succession during infancy.
Collapse
Affiliation(s)
- Candice Quin
- Department of Biology, University of British Columbia, Kelowna, Canada
| | - Deanna L. Gibson
- Department of Biology, University of British Columbia, Kelowna, Canada,Department of Medicine, University of British Columbia, Kelowna, Canada,CONTACT Deanna L. Gibson Department of Biology, University of British Columbia, Okanagan Campus, ASC 386, 3187 University Way, Kelowna, BCV1V 1V7, Canada
| |
Collapse
|
46
|
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: 7] [Impact Index Per Article: 1.4] [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
|
47
|
Brushett S, Sinha T, Reijneveld SA, de Kroon MLA, Zhernakova A. The Effects of Urbanization on the Infant Gut Microbiota and Health Outcomes. Front Pediatr 2020; 8:408. [PMID: 32903831 PMCID: PMC7438894 DOI: 10.3389/fped.2020.00408] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/12/2020] [Indexed: 12/14/2022] Open
Abstract
Humans and their gut microbiota have co-evolved over thousands of years, resulting in the establishment of a complex host-microbiota ecosystem. Early life environmental factors, such as delivery mode, nutrition, and medication use, have been shown to substantially affect both host-microbiota interactions and health outcomes. However, the effects of urbanization (characterized by the spectrum of rural and urban populations) on these early life events have been overlooked. A deeper understanding of the relationship between urbanization and microbiota development will allow for the identification of novel biological and social approaches that can be implemented to prevent and treat disease and promote maternal and infant/child health. The aim of this narrative review is to summarize how factors associated with urbanization differentially impact delivery mode, nutrition, and medication use, and how these changes subsequently affect the gut microbiota and health outcomes of infants. This narrative review also describes the important evidence gaps associated with these relationships and recommends actions that can be taken to improve the health of mothers and infants worldwide.
Collapse
Affiliation(s)
- Siobhan Brushett
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Health Sciences, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Trishla Sinha
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Sijmen A. Reijneveld
- Department of Health Sciences, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Marlou L. A. de Kroon
- Department of Health Sciences, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
48
|
LeMay-Nedjelski L, Butcher J, Ley SH, Asbury MR, Hanley AJ, Kiss A, Unger S, Copeland JK, Wang PW, Zinman B, Stintzi A, O'Connor DL. Examining the relationship between maternal body size, gestational glucose tolerance status, mode of delivery and ethnicity on human milk microbiota at three months post-partum. BMC Microbiol 2020; 20:219. [PMID: 32689933 PMCID: PMC7372813 DOI: 10.1186/s12866-020-01901-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 07/13/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Few studies have examined how maternal body mass index (BMI), mode of delivery and ethnicity affect the microbial composition of human milk and none have examined associations with maternal metabolic status. Given the high prevalence of maternal adiposity and impaired glucose metabolism, we systematically investigated the associations between these maternal factors in women ≥20 years and milk microbial composition and predicted functionality by V4-16S ribosomal RNA gene sequencing (NCT01405547; https://clinicaltrials.gov/ct2/show/NCT01405547 ). Demographic data, weight, height, and a 3-h oral glucose tolerance test were gathered at 30 (95% CI: 25-33) weeks gestation, and milk samples were collected at 3 months post-partum (n = 113). RESULTS Multivariable linear regression analyses demonstrated no significant associations between maternal characteristics (maternal BMI [pre-pregnancy, 3 months post-partum], glucose tolerance, mode of delivery and ethnicity) and milk microbiota alpha-diversity; however, pre-pregnancy BMI was associated with human milk microbiota beta-diversity (Bray-Curtis R2 = 0.037). Women with a pre-pregnancy BMI > 30 kg/m2 (obese) had a greater incidence of Bacteroidetes (incidence rate ratio [IRR]: 3.70 [95% CI: 1.61-8.48]) and a reduced incidence of Proteobacteria (0.62 [0.43-0.90]) in their milk, compared to women with an overweight BMI (25.0-29.9 kg/m2) as assessed by multivariable Poisson regression. An increased incidence of Gemella was observed among mothers with gestational diabetes who had an overweight BMI versus healthy range BMI (5.96 [1.85-19.21]). An increased incidence of Gemella was also observed among mothers with impaired glucose tolerance with an obese BMI versus mothers with a healthy range BMI (4.04 [1.63-10.01]). An increased incidence of Brevundimonas (16.70 [5.99-46.57]) was found in the milk of women who underwent an unscheduled C-section versus vaginal delivery. Lastly, functional gene inference demonstrated that pre-pregnancy obesity was associated with an increased abundance of genes encoding for the biosynthesis of secondary metabolites pathway in milk (coefficient = 0.0024, PFDR < 0.1). CONCLUSIONS Human milk has a diverse microbiota of which its diversity and differential abundance appear associated with maternal BMI, glucose tolerance status, mode of delivery, and ethnicity. Further research is warranted to determine whether this variability in the milk microbiota impacts colonization of the infant gut.
Collapse
Affiliation(s)
- Lauren LeMay-Nedjelski
- Department of Nutritional Sciences, University of Toronto, Medical Sciences Building, 1 King College Circle, Toronto, ON, M5S 1A8, Canada.,Peter Gilgan Centre for Research and Learning, Translational Medicine, The Hospital for Sick Children, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
| | - James Butcher
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Sylvia H Ley
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, 1440 Canal Street, Suite 2001, Mail Box 8318, New Orleans, LA, 70112, USA
| | - Michelle R Asbury
- Department of Nutritional Sciences, University of Toronto, Medical Sciences Building, 1 King College Circle, Toronto, ON, M5S 1A8, Canada.,Peter Gilgan Centre for Research and Learning, Translational Medicine, The Hospital for Sick Children, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
| | - Anthony J Hanley
- Department of Nutritional Sciences, University of Toronto, Medical Sciences Building, 1 King College Circle, Toronto, ON, M5S 1A8, Canada
| | - Alex Kiss
- Department of Research Design and Biostatistics, Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON, M4N 3M5, Canada
| | - Sharon Unger
- Department of Nutritional Sciences, University of Toronto, Medical Sciences Building, 1 King College Circle, Toronto, ON, M5S 1A8, Canada.,Department of Pediatrics, Mount Sinai Hospital, 600 University Ave, Toronto, ON, M5G 1X5, Canada.,Department of Pediatrics, University of Toronto, Medical Sciences Building, 1 King College Cir, Toronto, ON, M5S 1A8, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, 600 University Ave, Toronto, ON, M5G 1X5, Canada
| | - Julia K Copeland
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, 25 Willcocks Street, Toronto, ON, M5S 3B2, Canada
| | - Pauline W Wang
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, 25 Willcocks Street, Toronto, ON, M5S 3B2, Canada
| | - Bernard Zinman
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, 25 Willcocks Street, Toronto, ON, M5S 3B2, Canada
| | - Alain Stintzi
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Deborah L O'Connor
- Department of Nutritional Sciences, University of Toronto, Medical Sciences Building, 1 King College Circle, Toronto, ON, M5S 1A8, Canada. .,Peter Gilgan Centre for Research and Learning, Translational Medicine, The Hospital for Sick Children, 686 Bay Street, Toronto, ON, M5G 0A4, Canada. .,Department of Pediatrics, Mount Sinai Hospital, 600 University Ave, Toronto, ON, M5G 1X5, Canada.
| |
Collapse
|
49
|
Corona-Cervantes K, García-González I, Villalobos-Flores LE, Hernández-Quiroz F, Piña-Escobedo A, Hoyo-Vadillo C, Rangel-Calvillo MN, García-Mena J. Human milk microbiota associated with early colonization of the neonatal gut in Mexican newborns. PeerJ 2020; 8:e9205. [PMID: 32509465 PMCID: PMC7247532 DOI: 10.7717/peerj.9205] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 04/25/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Human milk microbiota plays a role in the bacterial colonization of the neonatal gut, which has important consequences in the health and development of the newborn. However, there are few studies about the vertical transfer of bacteria from mother to infant in Latin American populations. METHODS We performed a cross-sectional study characterizing the bacterial diversity of 67 human milk-neonatal stool pairs by high-throughput sequencing of V3-16S rDNA libraries, to assess the effect of the human milk microbiota on the bacterial composition of the neonate's gut at early days. RESULTS Human milk showed higher microbial diversity as compared to the neonatal stool. Members of the Staphylococcaceae and Sphingomonadaceae families were more prevalent in human milk, whereas the Pseudomonadaceae family, Clostridium and Bifidobacterium genera were in the neonatal stool. The delivery mode showed association with the neonatal gut microbiota diversity, but not with the human milk microbiota diversity; for instance, neonates born by C-section showed greater richness and diversity in stool microbiota than those born vaginally. We found 25 bacterial taxa shared by both ecosystems and 67.7% of bacteria found in neonate stool were predicted to originate from human milk. This study contributes to the knowledge of human milk and neonatal stool microbiota in healthy Mexican population and supports the idea of vertical mother-neonate transmission through exclusive breastfeeding.
Collapse
Affiliation(s)
- Karina Corona-Cervantes
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, CDMX, Mexico
| | - Igrid García-González
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, CDMX, Mexico
| | - Loan Edel Villalobos-Flores
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, CDMX, Mexico
| | - Fernando Hernández-Quiroz
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, CDMX, Mexico
| | - Alberto Piña-Escobedo
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, CDMX, Mexico
| | - Carlos Hoyo-Vadillo
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, CDMX, Mexico
| | - Martín Noé Rangel-Calvillo
- Hospital General “Dr. José María Rodríguez”, Instituto de Salud del Estado de México, Ecatepec de Morelos, Estado de Mexico, Mexico
| | - Jaime García-Mena
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, CDMX, Mexico
| |
Collapse
|
50
|
Angelopoulou A, Warda AK, O'Connor PM, Stockdale SR, Shkoporov AN, Field D, Draper LA, Stanton C, Hill C, Ross RP. Diverse Bacteriocins Produced by Strains From the Human Milk Microbiota. Front Microbiol 2020; 11:788. [PMID: 32508758 PMCID: PMC7248182 DOI: 10.3389/fmicb.2020.00788] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 04/02/2020] [Indexed: 12/16/2022] Open
Abstract
Microbial colonization of the infant gut is a convoluted process dependent on numerous contributing factors, including age, mode of delivery and diet among others that has lifelong implication for human health. Breast milk also contains a microbiome which acts as a source of colonizing bacteria for the infant. Here, we demonstrate that human milk harbors a wide diversity of bacteriocin-producing strains with the potential to compete among the developing gut microbiota of the infant. We screened 37 human milk samples and found isolates with antimicrobial activity and distinct cross-immunity profiles. From these isolates, we detected 73 putative gene clusters for bacteriocins of all known sub-classes, including 16 novel prepeptides. More specifically, we detected two novel lantibiotics, four sactibiotics and three class IIa bacteriocins with an unusual modification of the pediocin box that is composed of YDNGI instead of the highly conserved motif YGNGV. Moreover, we identified a novel class IIb bacteriocin, four novel class IIc and two class IId bacteriocins. In conclusion, human milk contains a variety of bacteriocin-producing strains which may provide them a competitive advantage in the colonization of the infant gut and suggests that the milk microbiota is a source of antimicrobial potential.
Collapse
Affiliation(s)
- Angeliki Angelopoulou
- School of Microbiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Alicja K Warda
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Paula M O'Connor
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
| | | | | | - Des Field
- School of Microbiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | | | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
| | - Colin Hill
- School of Microbiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - R Paul Ross
- School of Microbiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
| |
Collapse
|