1
|
Li F, Hooi SL, Choo YM, Teh CSJ, Toh KY, Lim LWZ, Lee YQ, Chong CW, Ahmad Kamar A. Progression of gut microbiome in preterm infants during the first three months. Sci Rep 2025; 15:12104. [PMID: 40204761 PMCID: PMC11982265 DOI: 10.1038/s41598-025-95198-1] [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: 06/05/2024] [Accepted: 03/19/2025] [Indexed: 04/11/2025] Open
Abstract
The colonization and evolution of gut microbiota early in life play a vital role in shaping a healthy, robust immune system for infant health, whether in combating short-term illness or improving long-term health outcomes. Early-life clinical practices may interrupt or disrupt the normal colonization process of the infant gut microbiota, thereby increasing disease susceptibility. In this prospective cohort study, we analyzed the gut microbiota of 46 term and 23 preterm infants using 16S rRNA gene metagenomic sequencing. Fecal samples were collected at six timepoints during the first three months of life. Notably, gestational age was the main factor contributing to differences in the meconium microbial composition. Intriguingly, our study unveiled a more homogeneous microbial composition in preterm infants with more abundant Bifidobacterium from the postnatal age (PNA) of one month. Concurrently, the beneficial bacteria Bifidobacterium and Lactobacillus gradually increased, and the potentially pathogenic bacteria Clostridium, Enterobacter, Enterococcus, Klebsiella, and Pseudomonas gradually decreased. Furthermore, our study underscored a link between decreased microbial diversity of preterm infants and exclusive breastfeeding and antibiotic exposure. Moreover, preterm infants with patent ductus arteriosus (PDA) exhibited reduced microbial diversity but higher abundances of Streptococcus oralis and Streptococcus mitis.
Collapse
Affiliation(s)
- Fangfang Li
- Department of Pediatrics, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | | | - Yao Mun Choo
- Department of Pediatrics, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
| | - Cindy Shuan Ju Teh
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
| | | | | | - Yee Qing Lee
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Chun Wie Chong
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, 47500, Selangor, Malaysia
- Monash Microbiome Research Centre, Monash University Malaysia, Bandar Sunway, 47500, Selangor, Malaysia
| | - Azanna Ahmad Kamar
- Department of Pediatrics, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| |
Collapse
|
2
|
Reuben RC, Torres C. Integrating the milk microbiome signatures in mastitis: milk-omics and functional implications. World J Microbiol Biotechnol 2025; 41:41. [PMID: 39826029 PMCID: PMC11742929 DOI: 10.1007/s11274-024-04242-1] [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: 10/13/2024] [Accepted: 12/26/2024] [Indexed: 01/20/2025]
Abstract
Mammalian milk contains a variety of complex bioactive and nutritional components and microorganisms. These microorganisms have diverse compositions and functional roles that impact host health and disease pathophysiology, especially mastitis. The advent and use of high throughput omics technologies, including metagenomics, metatranscriptomics, metaproteomics, metametabolomics, as well as culturomics in milk microbiome studies suggest strong relationships between host phenotype and milk microbiome signatures in mastitis. While single omics studies have undoubtedly contributed to our current understanding of milk microbiome and mastitis, they often provide limited information, targeting only a single biological viewpoint which is insufficient to provide system-wide information necessary for elucidating the biological footprints and molecular mechanisms driving mastitis and milk microbiome dysbiosis. Therefore, integrating a multi-omics approach in milk microbiome research could generate new knowledge, improve the current understanding of the functional and structural signatures of the milk ecosystem, and provide insights for sustainable mastitis control and microbiome management.
Collapse
Affiliation(s)
- Rine Christopher Reuben
- Biology Department, King's College, 133 North River Street, Wilkes-Barre, PA, 18711, USA.
- Area of Biochemistry and Molecular Biology, OneHealth-UR Research Group, University of La Rioja, 26006, Logroño, Spain.
| | - Carmen Torres
- Area of Biochemistry and Molecular Biology, OneHealth-UR Research Group, University of La Rioja, 26006, Logroño, Spain
| |
Collapse
|
3
|
Ma J, Palmer DJ, Geddes D, Lai CT, Rea A, Prescott SL, D'Vaz N, Stinson LF. Maternal Allergic Disease Phenotype and Infant Birth Season Influence the Human Milk Microbiome. Allergy 2024. [PMID: 39723602 DOI: 10.1111/all.16442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 10/11/2024] [Accepted: 11/19/2024] [Indexed: 12/28/2024]
Abstract
Early infancy is a critical period for immune development. In addition to being the primary food source during early infancy, human milk also provides multiple bioactive components that shape the infant gut microbiome and immune system and provides a constant source of exposure to maternal microbiota. Given the potential interplay between allergic diseases and the human microbiome, this study aimed to characterise the milk microbiome of allergic mothers. Full-length 16S rRNA gene sequencing was performed on milk samples collected at 3 and 6 months postpartum from 196 women with allergic disease. Multivariate linear mixed models were constructed to identify the maternal, infant, and environmental determinants of the milk microbiome. Human milk microbiome composition and beta diversity varied over time (PERMANOVA R2 = 0.011, p = 0.011). The season of infant birth emerged as the strongest determinant of the microbiome community structure (PERMANOVA R2 = 0.014, p = 0.011) with impacts on five of the most abundant taxa. The milk microbiome also varied according to the type of maternal allergic disease (allergic rhinitis, asthma, atopic dermatitis, and food allergy). Additionally, infant formula exposure reduced the relative abundance of several typical oral taxa in milk. In conclusion, the milk microbiome of allergic mothers was strongly shaped by the season of infant birth, maternal allergic disease phenotype, and infant feeding mode. Maternal allergic disease history and infant season of birth should therefore be considered in future studies of infant and maternal microbiota. Trial Registration: ClinicalTrials.gov identifier: ACTRN12606000281594.
Collapse
Affiliation(s)
- Jie Ma
- School of Molecular Sciences, The University of Western Australia, Crawley, WA, Australia
- ABREAST Network, Perth, WA, Australia
- UWA Centre for Human Lactation Research and Translation, Perth, WA, Australia
| | - Debra J Palmer
- ABREAST Network, Perth, WA, Australia
- Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
- School of Medicine, The University of Western Australia, Perth, WA, Australia
| | - Donna Geddes
- School of Molecular Sciences, The University of Western Australia, Crawley, WA, Australia
- ABREAST Network, Perth, WA, Australia
- UWA Centre for Human Lactation Research and Translation, Perth, WA, Australia
| | - Ching Tat Lai
- School of Molecular Sciences, The University of Western Australia, Crawley, WA, Australia
- ABREAST Network, Perth, WA, Australia
- UWA Centre for Human Lactation Research and Translation, Perth, WA, Australia
| | - Alethea Rea
- Mathematics and Statistics, Harry Butler Institute, Murdoch University, Murdoch, WA, Australia
| | - Susan L Prescott
- Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
- School of Medicine, The University of Western Australia, Perth, WA, Australia
- Nova Institute for Health, Baltimore, Maryland, USA
- Department of Family and Community Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Nina D'Vaz
- Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
- School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia
| | - Lisa F Stinson
- School of Molecular Sciences, The University of Western Australia, Crawley, WA, Australia
- ABREAST Network, Perth, WA, Australia
- UWA Centre for Human Lactation Research and Translation, Perth, WA, Australia
| |
Collapse
|
4
|
Amar Y, Grube J, Köberle M, Schaubeck M, Biedermann T, Volz T. Bifidobacterium breve DSM 32583 and Limosilactobacillus fermentum CECT5716 postbiotics attenuate S. aureus and IL-33-induced Th2 responses. Microbiol Res 2024; 289:127913. [PMID: 39316930 DOI: 10.1016/j.micres.2024.127913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/04/2024] [Accepted: 09/15/2024] [Indexed: 09/26/2024]
Abstract
Over the past decades, the prevalence of allergic diseases noticeably increased in industrialized countries. The Th2 immune response plays a central role in these pathologies and its modulation using pro-/postbiotics constitutes a promising approach to prevent or alleviate disease symptoms. The aim of this in vitro study, was to investigate the ability of human milk-derived Bifidobacterium breve DSM 32583 (Bb) and Limosilactobacillus fermentum CECT5716 (Lf), to modulate the Th2 induced responses. To this end, Th2 cells were generated by co-culturing of human naïve Th cells with monocyte-derived dendritic cells (moDCs) either stimulated with Staphylococcus aureus or IL-33. The immunomodulatory effects of pro-/postbiotic preparations of Bb and Lf on moDCs and Th2 cells were evaluated in terms of maturation markers expression and cytokines production. Remarkably, the tested strains induced the anti-inflammatory cytokine IL-10 in moDCs, in a strain-, dose- and viability-dependent manner with no significant upregulation of IL-12p70 nor CD83, CD86 or HLA-DR. Interestingly, Bb and Lf postbiotics were able to dampen the Th2/Th1 response induced upon S. aureus- or IL-33 stimulation. They were also able to synergistically induce IL-10 in moDCs and T cells, upon co-stimulation with LPS. Finally, we observed that live probiotics triggered a mild Th1 response that was attenuated in the presence of galacto-oligosaccharides. Altogether, Bb and Lf pro-/postbiotics exhibited remarkable immune regulatory effects on both moDCs and Th2 cells. Therefore, further in vivo studies should be considered to validate these findings and assess their ability to prevent allergy or alleviate its symptoms in affected patients.
Collapse
Affiliation(s)
- Yacine Amar
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich 80802, Germany.
| | - Jana Grube
- HiPP GmbH & Co. Vertrieb KG, Pfaffenhofen (Ilm) 85276, Germany
| | - Martin Köberle
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich 80802, Germany
| | | | - Tilo Biedermann
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich 80802, Germany
| | - Thomas Volz
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich 80802, Germany
| |
Collapse
|
5
|
Mariani Wigley ILC, Nazzari S, Pastore M, Grumi S, Provenzi L. Exclusive breastfeeding mitigates the association between prenatal maternal pandemic-related stress and children sleep problems at 24 months of age. Dev Psychopathol 2024:1-11. [PMID: 39397696 DOI: 10.1017/s0954579424001627] [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: 10/15/2024]
Abstract
Infant sleep quality is increasingly regarded as an important factor for children long-term functioning and adaptation. The early roots of sleep disturbances are still poorly understood and likely involve a complex interplay between prenatal and postnatal factors. This study investigated whether exclusive breastfeeding during the first 6 months moderated the association between maternal prenatal pandemic-related stress (PRS) and sleep problems in 24-months children born during the COVID-19 pandemic. We also explored the potential contribution of maternal postnatal anxiety in these relations. Seventy-eight infants (50% males) and their mothers provided complete data from birth to 24 months. Between 12 and 48 h from birth, maternal PRS during pregnancy was retrospectively reported as well as maternal anxiety and exclusive breastfeeding. Maternal anxiety and exclusive breastfeeding were also reported at 3 and 6 months after childbirth. Children sleep disturbances were reported at 24 months. Bayesian analyses revealed that maternal PRS was positively associated with sleep problems in children who were not exclusively breastfed from birth to 6 months. Findings add to the growing literature on the lasting impact of early pre- and postnatal experiences on child well-being and development.
Collapse
Affiliation(s)
- Isabella Lucia Chiara Mariani Wigley
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, Turku University Hospital and University of Turku, Turku, Finland
| | - Sarah Nazzari
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Massimiliano Pastore
- Department of Developmental and Social Psychology, University of Padua, Padova, Veneto, Italy
| | - Serena Grumi
- Developmental Psychobiology Lab, IRCCS Mondino Foundation, Pavia, Italy
| | - Livio Provenzi
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Developmental Psychobiology Lab, IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
6
|
Shao Y, Garcia-Mauriño C, Clare S, Dawson NJR, Mu A, Adoum A, Harcourt K, Liu J, Browne HP, Stares MD, Rodger A, Brocklehurst P, Field N, Lawley TD. Primary succession of Bifidobacteria drives pathogen resistance in neonatal microbiota assembly. Nat Microbiol 2024; 9:2570-2582. [PMID: 39242817 PMCID: PMC11445081 DOI: 10.1038/s41564-024-01804-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 08/05/2024] [Indexed: 09/09/2024]
Abstract
Human microbiota assembly commences at birth, seeded by both maternal and environmental microorganisms. Ecological theory postulates that primary colonizers dictate microbial community assembly outcomes, yet such microbial priority effects in the human gut remain underexplored. Here using longitudinal faecal metagenomics, we characterized neonatal microbiota assembly for a cohort of 1,288 neonates from the UK. We show that the pioneering neonatal gut microbiota can be stratified into one of three distinct community states, each dominated by a single microbial species and influenced by clinical and host factors, such as maternal age, ethnicity and parity. A community state dominated by Enterococcus faecalis displayed stochastic microbiota assembly with persistent high pathogen loads into infancy. In contrast, community states dominated by Bifidobacterium, specifically B. longum and particularly B. breve, exhibited a stable assembly trajectory and long-term pathogen colonization resistance, probably due to strain-specific functional adaptions to a breast milk-rich neonatal diet. Consistent with our human cohort observation, B. breve demonstrated priority effects and conferred pathogen colonization resistance in a germ-free mouse model. Our findings solidify the crucial role of Bifidobacteria as primary colonizers in shaping the microbiota assembly and functions in early life.
Collapse
Affiliation(s)
- Yan Shao
- Host-Microbiota Interactions Laboratory, Wellcome Sanger Institute, Hinxton, UK.
| | | | - Simon Clare
- Host-Microbiota Interactions Laboratory, Wellcome Sanger Institute, Hinxton, UK
| | - Nicholas J R Dawson
- Host-Microbiota Interactions Laboratory, Wellcome Sanger Institute, Hinxton, UK
| | - Andre Mu
- Host-Microbiota Interactions Laboratory, Wellcome Sanger Institute, Hinxton, UK
| | - Anne Adoum
- Host-Microbiota Interactions Laboratory, Wellcome Sanger Institute, Hinxton, UK
| | - Katherine Harcourt
- Host-Microbiota Interactions Laboratory, Wellcome Sanger Institute, Hinxton, UK
| | - Junyan Liu
- Host-Microbiota Interactions Laboratory, Wellcome Sanger Institute, Hinxton, UK
| | - Hilary P Browne
- Host-Microbiota Interactions Laboratory, Wellcome Sanger Institute, Hinxton, UK
| | - Mark D Stares
- Host-Microbiota Interactions Laboratory, Wellcome Sanger Institute, Hinxton, UK
| | - Alison Rodger
- Institute for Global Health, University College London, London, UK
| | - Peter Brocklehurst
- Birmingham Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Nigel Field
- Institute for Global Health, University College London, London, UK
| | - Trevor D Lawley
- Host-Microbiota Interactions Laboratory, Wellcome Sanger Institute, Hinxton, UK.
| |
Collapse
|
7
|
Tobin NH, Li F, Brummel S, Flynn PM, Dababhai S, Moodley D, Chinula L, Violari A, Fowler MG, Rouzier V, Kuhn L, Aldrovandi GM. Maternal HIV infection and the milk microbiome. MICROBIOME 2024; 12:182. [PMID: 39342403 PMCID: PMC11439335 DOI: 10.1186/s40168-024-01843-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/21/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Children born to women with HIV but who do not become HIV infected experience increased morbidity and mortality compared with children born to women without HIV. The basis of this increased vulnerability is unknown. The microbiome, specifically the infant gut microbiome, likely plays an important role in infant immune development. The human milk microbiome is thought to have an important role in the development of the infant gut and therefore, if perturbed, may contribute to this increased vulnerability. We investigated the effects of HIV and its therapies on the milk microbiome and possible changes in the milk microbiome before or after infant HIV infection. RESULTS Seven-hundred fifty-six human milk samples were selected from three separate studies conducted over a 15-year period to investigate the role of HIV and its therapies on the human milk microbiome. Our data reveal that the milk microbiome is modulated by parity (R2 = 0.006, p = 0.041), region/country (R2 = 0.014, p = 0.007), and duration of lactation (R2 = 0.027-0.038, all p < 0.001). There is no evidence, however, using 16S rRNA V4 amplicon sequencing, that the human milk microbiome is altered by HIV infection (R2 = 0.003, p = 0.896), by combination antiretroviral therapy (R2 = 0.0009, p = 0.909), by advanced maternal disease (R2 = 0.003, p = 0.263), or in cases of infant infection either through isolated early mucosal (R2 = 0.003, p = 0.197) or early mucosal and breast milk transmission (R2 = 0.002, p = 0.587). CONCLUSIONS The milk microbiome varies by stage of lactation, by parity, and by region; however, we found no evidence that the human milk microbiome is altered by maternal HIV infection, disease severity, or antiretroviral therapy. Additionally, we found no association between the milk microbiome and transmission of HIV to the infant. Investigations including higher resolution microbiome approaches or into other potential mechanisms to understand why the approximately one million children born annually to women with HIV escape infection, but do not escape harm, are urgently needed. Video Abstract.
Collapse
Affiliation(s)
- Nicole H Tobin
- Division of Infectious Diseases, Department of Pediatrics, David Geffen School of Medicine at the University of California, 10833 Le Conte Ave., 22-340 MDCC, Los Angeles, CA, 90095, USA
| | - Fan Li
- Division of Infectious Diseases, Department of Pediatrics, David Geffen School of Medicine at the University of California, 10833 Le Conte Ave., 22-340 MDCC, Los Angeles, CA, 90095, USA
| | - Sean Brummel
- The Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Patricia M Flynn
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sufia Dababhai
- Johns Hopkins Bloomberg School of Public Health, Blantyre, Malawi
| | - Dhayendre Moodley
- Centre for the AIDS Programme of Research, School of Clinical Medicine, University of KwaZulu Natal, Durban, South Africa
| | - Lameck Chinula
- University of North Carolina (UNC) Project Malawi, Lilongwe, Malawi
| | - Avy Violari
- Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Mary Glenn Fowler
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vanessa Rouzier
- The Haitian Group for the Study of Kaposi's Sarcoma and Opportunistic Infections, GHESKIO Centers, Port-Au-Price, Ouest, 15727, Haiti
| | - Louise Kuhn
- Gertrude H. Sergievsky Center, Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY, USA
| | - Grace M Aldrovandi
- Division of Infectious Diseases, Department of Pediatrics, David Geffen School of Medicine at the University of California, 10833 Le Conte Ave., 22-340 MDCC, Los Angeles, CA, 90095, USA.
| |
Collapse
|
8
|
Shama S, Asbury MR, Kiss A, Bando N, Butcher J, Comelli EM, Copeland JK, Greco A, Kothari A, Sherman PM, Stintzi A, Taibi A, Tomlinson C, Unger S, Wang PW, O'Connor DL. Mother's milk microbiota is associated with the developing gut microbial consortia in very-low-birth-weight infants. Cell Rep Med 2024; 5:101729. [PMID: 39243753 PMCID: PMC11525026 DOI: 10.1016/j.xcrm.2024.101729] [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: 04/16/2024] [Revised: 06/30/2024] [Accepted: 08/16/2024] [Indexed: 09/09/2024]
Abstract
Mother's milk contains diverse bacterial communities, although their impact on microbial colonization in very-low-birth-weight (VLBW, <1,500 g) infants remains unknown. Here, we examine relationships between the microbiota in preterm mother's milk and the VLBW infant gut across initial hospitalization (n = 94 mother-infant dyads, 422 milk-stool pairs). Shared zero-radius operational taxonomic units (zOTUs) between milk-stool pairs account for ∼30%-40% of zOTUs in the VLBW infant's gut. We show dose-response relationships between intakes of several genera from milk and their concentrations in the infant's gut. These relationships and those related to microbial sharing change temporally and are modified by in-hospital feeding practices (especially direct breastfeeding) and maternal-infant antibiotic use. Correlations also exist between milk and stool microbial consortia, suggesting that multiple milk microbes may influence overall gut communities together. These results highlight that the mother's milk microbiota may shape the gut colonization of VLBW infants by delivering specific bacteria and through intricate microbial interactions.
Collapse
Affiliation(s)
- Sara Shama
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada; Translational Medicine Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
| | - Michelle R Asbury
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada; Departments of Physiology & Pharmacology, and Pediatrics, University of Calgary, Calgary, AB, Canada
| | - Alex Kiss
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada; Evaluative and Clinical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Nicole Bando
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada; Translational Medicine Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
| | - James Butcher
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada; Ottawa Institute of Systems Biology, Ottawa, ON, Canada
| | - Elena M Comelli
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada; Joannah and Brian Lawson Centre for Child Nutrition, University of Toronto, Toronto, ON, Canada
| | - Julia K Copeland
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, ON, Canada
| | - Adrianna Greco
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada; Translational Medicine Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
| | - Akash Kothari
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada; Translational Medicine Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
| | - Philip M Sherman
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada; Cell Biology Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, ON, Canada; Gastroenterology, Hepatology, and Nutrition, The Hospital for Sick Children, Toronto, ON, Canada; Department of Paediatrics, University of Toronto, Toronto, ON, Canada
| | - Alain Stintzi
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada; Ottawa Institute of Systems Biology, Ottawa, ON, Canada
| | - Amel Taibi
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - Christopher Tomlinson
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada; Translational Medicine Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, ON, Canada; Joannah and Brian Lawson Centre for Child Nutrition, University of Toronto, Toronto, ON, Canada; Department of Paediatrics, University of Toronto, Toronto, ON, Canada; Division of Neonatology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Sharon Unger
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada; Department of Paediatrics, University of Toronto, Toronto, ON, Canada; Division of Neonatology, Izaak Walton Killam Hospital, Halifax, NS, Canada; Department of Pediatrics, Sinai Health, Toronto, ON, Canada; Rogers Hixon Ontario Human Milk Bank, Toronto, ON, Canada
| | - Pauline W Wang
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, ON, Canada
| | - Deborah L O'Connor
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada; Translational Medicine Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, ON, Canada; Joannah and Brian Lawson Centre for Child Nutrition, University of Toronto, Toronto, ON, Canada; Department of Pediatrics, Sinai Health, Toronto, ON, Canada; Rogers Hixon Ontario Human Milk Bank, Toronto, ON, Canada.
| |
Collapse
|
9
|
Lee HY, Nazmul T, Lan J, Oyoshi MK. Maternal influences on offspring food allergy. Immunol Rev 2024; 326:130-150. [PMID: 39275992 PMCID: PMC11867100 DOI: 10.1111/imr.13392] [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: 09/16/2024]
Abstract
The prevalence of allergies has been globally escalating. While allergies could appear at any age, they often develop in early life. However, the significant knowledge gap in the field is the mechanisms by which allergies affect certain people but not others. Investigating early factors and events in neonatal life that have a lasting impact on determining the susceptibilities of children to develop allergies is a significant area of the investigation as it promotes the understanding of neonatal immune system that mediates tolerance versus allergies. This review focuses on the research over the recent 10 years regarding the potential maternal factors that influence offspring allergies with a view to food allergy, a potentially life-threatening cause of anaphylaxis. The role of breast milk, maternal diet, maternal antibodies, and microbiota that have been suggested as key maternal factors regulating offspring allergies are discussed here. We also suggest future research area to expand our knowledge of maternal-offspring interactions on the pathogenesis of food allergy.
Collapse
Affiliation(s)
- Hwa Yeong Lee
- Division of Pediatric Allergy, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Charlestown, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Tanuza Nazmul
- Division of Pediatric Allergy, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Charlestown, MA
| | - Jinggang Lan
- Division of Pediatric Allergy, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Charlestown, MA
| | - Michiko K. Oyoshi
- Division of Pediatric Allergy, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Charlestown, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
10
|
Ingram K, Gregg C, Tegge A, Elison JT, Lin W, Howell BR. Metagenomic assessment of the bacterial breastfeeding microbiome in mature milk across lactation. Front Pediatr 2024; 11:1275436. [PMID: 39092171 PMCID: PMC11292495 DOI: 10.3389/fped.2023.1275436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/25/2023] [Indexed: 08/04/2024] Open
Abstract
Introduction Research has illustrated the presence of a diverse range of microbiota in human milk. The composition of the milk microbiome varies across different stages of lactation, emphasizing the need to consider the lactation stage when studying its composition. Additionally, the transfer of both milk and skin microbiota during breastfeeding is crucial for understanding their collective impact on infant health and development. Further exploration of the complete breastfeeding microbiome is necessary to unravel the role these organisms play in infant development. We aim to longitudinally assess the bacterial breastfeeding microbiome across stages of lactation. This includes all the bacteria that infants are exposed to during breastfeeding, such as bacteria found within human milk and any bacteria found on the breast and nipple. Methods Forty-six human milk samples were collected from 15 women at 1, 4, 7, and 10 months postpartum. Metagenomic analysis of the bacterial microbiome for these samples was performed by CosmosID (Rockville, MD) via deep sequencing. Results Staphylococcus epidermidis and Propionibacteriaceae species are the most abundant bacterial species from these samples. Samples collected at 10 months showed higher abundances of Proteobacteria, Streptococcaceae, Lactobacillales, Streptococcus, and Neisseria mucosa compared to other timepoints. Alpha diversity varied greatly between participants but did not change significantly over time. Discussion As the bacterial breastfeeding microbiome continues to be studied, bacterial contributions could be used to predict and reduce health risks, optimize infant outcomes, and design effective management strategies, such as altering the maternal flora, to mitigate adverse health concerns.
Collapse
Affiliation(s)
- Kelly Ingram
- Virginia Tech Carilion School of Medicine, Roanoke, VA, United States
| | - Collin Gregg
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, United States
| | - Allison Tegge
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, United States
| | - Jed T. Elison
- Institute for Child Development, University of Minnesota, Minneapolis, MN, United States
- Masonic Institute for the Developing Brain, University of Minnesota, St. Paul, MN, United States
| | - Weili Lin
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Brittany R. Howell
- Virginia Tech Carilion School of Medicine, Roanoke, VA, United States
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, United States
- Department of Human Development and Family Science, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
11
|
Tsifintaris M, Sitmalidis M, Tokamani M, Anastasiadi C, Georganta M, Tsochantaridis I, Vlachakis D, Tsikouras P, Nikolettos N, Chrousos GP, Sandaltzopoulos R, Giannakakis A. Analysis of Human Milk Microbiota in Northern Greece by Comparative 16S rRNA Sequencing vs. Local Dairy Animals. Nutrients 2024; 16:2175. [PMID: 39064618 PMCID: PMC11280067 DOI: 10.3390/nu16142175] [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: 06/06/2024] [Revised: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Milk is a biological fluid with a dynamic composition of micronutrients and bioactive molecules that serves as a vital nutrient source for infants. Milk composition is affected by multiple factors, including genetics, geographical location, environmental conditions, lactation phase, and maternal nutrition, and plays a key role in dictating its microbiome. This study addresses a less-explored aspect, comparing the microbial communities in human breast milk with those in mature milk from species that are used for milk consumption. Since mature animal milk is used as a supplement for both the infant (formula) and the child/adolescent, our main aim was to identify shared microbial communities in colostrum and mature human milk. Using 16S rRNA metagenomic sequencing, we focused on characterizing the milk microbiota in the Northern Greek population by identifying shared microbial communities across samples and comparing the relative abundance of prevalent genera. We analyzed ten human milk samples (from five mothers), with five collected three days postpartum (colostrum) and five collected thirty to forty days postpartum (mature milk) from corresponding mothers. To perform an interspecies comparison of human milk microbiota, we analyzed five goat and five bovine milk samples from a local dairy industry, collected fifty to seventy days after birth. Alpha diversity analysis indicated moderate diversity and stability in bovine milk, high richness in goat milk, and constrained diversity in breast milk. Beta diversity analysis revealed significant distinctions among mammalian species, emphasizing both presence/absence and abundance-based clustering. Despite noticeable differences, shared microbial components underscore fundamental aspects across all mammalian species, highlighting the presence of a core microbiota predominantly comprising the Proteobacteria, Firmicutes, and Actinobacteriota phyla. At the genus level, Acinetobacter, Gemella, and Sphingobium exhibit significant higher abundance in human milk compared to bovine and goat milk, while Pseudomonas and Atopostipes are more prevalent in animal milk. Our comparative analysis revealed differences and commonalities in the microbial communities of various mammalian milks and unraveled the existence of a common fundamental milk core microbiome. We thus revealed both species-specific and conserved microbial communities in human, bovine, and goat milk. The existence of a common core microbiome with conserved differences between colostrum and mature human milk underscores fundamental similarities in the microbiota of milk across mammalian species, which could offer valuable implications for optimizing the nutritional quality and safety of dairy products as well as supplements for infant health.
Collapse
Affiliation(s)
- Margaritis Tsifintaris
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (M.T.)
| | - Michail Sitmalidis
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (M.T.)
| | - Maria Tokamani
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (M.T.)
| | - Christina Anastasiadi
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (M.T.)
| | - Maria Georganta
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Ilias Tsochantaridis
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (M.T.)
| | - Dimitrios Vlachakis
- Department of Biotechnology, Agricultural University of Athens, 11855 Athens, Greece
| | - Panagiotis Tsikouras
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Nikolaos Nikolettos
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - George P. Chrousos
- University Research Institute of Maternal and Child Health and Precision Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
- UNESCO Chair of Adolescent Health, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Raphael Sandaltzopoulos
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (M.T.)
| | - Antonis Giannakakis
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (M.T.)
| |
Collapse
|
12
|
Sun Q, Zhou Q, Ge S, Liu L, Li P, Gu Q. Effects of Maternal Diet on Infant Health: A Review Based on Entero-Mammary Pathway of Intestinal Microbiota. Mol Nutr Food Res 2024; 68:e2400077. [PMID: 39059011 DOI: 10.1002/mnfr.202400077] [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: 01/31/2024] [Revised: 04/30/2024] [Indexed: 07/28/2024]
Abstract
SCOPE The microbes in breast milk are critical for the early establishment of infant gut microbiota and have important implications for infant health. Breast milk microbes primarily derive from the migration of maternal intestinal microbiota. This review suggests that the regulation of maternal diet on gut microbiota may be an effective strategy to improve infant health. METHODS AND RESULTS This article reviews the impact of breast milk microbiota on infant development and intestinal health. The close relationship between the microbiota in the maternal gut and breast through the entero-mammary pathway is discussed. Based on the effect of diet on gut microbiota, it is proposed that changing the maternal dietary structure is a new strategy for regulating breast milk microbiota and infant intestinal microbiota, which would have a positive impact on infant health. CONCLUSION Breast milk microbes have beneficial effects on infant development and regulation of the immune system. The mother's gut and breast can undergo certain bacterial migration through the entero-mammary pathway. Research has shown that intervening in a mother's diet during breastfeeding can affect the composition of the mother's gut microbiota, thereby regulating the microbiota of breast milk and infant intestines, and is closely related to infant health.
Collapse
Affiliation(s)
- Qiaoyu Sun
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Qingqing Zhou
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Sitong Ge
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Lingli Liu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Ping Li
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Qing Gu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China
| |
Collapse
|
13
|
Inchingolo F, Inchingolo AM, Latini G, Ferrante L, de Ruvo E, Campanelli M, Longo M, Palermo A, Inchingolo AD, Dipalma G. Difference in the Intestinal Microbiota between Breastfeed Infants and Infants Fed with Artificial Milk: A Systematic Review. Pathogens 2024; 13:533. [PMID: 39057760 PMCID: PMC11280328 DOI: 10.3390/pathogens13070533] [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: 05/29/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
The gut microbiota (GM) plays a crucial role in human health, particularly during the first years of life. Differences in GM between breastfed and formula (F)-fed infants may influence long-term health outcomes. This systematic review aims to compare the gut microbiota of breastfed infants with that of F-fed infants and to evaluate the clinical implications of these differences. We searched databases on Scopus, Web of Science, and Pubmed with the following keywords: "gut microbiota", "gut microbiome", and "neonatal milk". The inclusion criteria were articles relating to the analysis of the intestinal microbiome of newborns in relation to the type of nutrition, clinical studies or case series, excluding reviews, meta-analyses, animal models, and in vitro studies. The screening phase ended with the selection of 13 publications for this work. Breastfed infants showed higher levels of beneficial bacteria such as Bifidobacterium and Lactobacillus, while F-fed infants had a higher prevalence of potentially pathogenic bacteria, including Clostridium difficile and Enterobacteriaceae. Infant feeding type influences the composition of oral GM significantly. Breastfeeding promotes a healthier and more diverse microbial ecosystem, which may offer protective health benefits. Future research should explore strategies to improve the GM of F-fed infants and understand the long-term health implications.
Collapse
Affiliation(s)
- Francesco Inchingolo
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy or (A.M.I.); or (G.L.); or (L.F.); or (E.d.R.); (M.L.); or (A.D.I.); or (G.D.)
| | - Angelo Michele Inchingolo
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy or (A.M.I.); or (G.L.); or (L.F.); or (E.d.R.); (M.L.); or (A.D.I.); or (G.D.)
| | - Giulia Latini
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy or (A.M.I.); or (G.L.); or (L.F.); or (E.d.R.); (M.L.); or (A.D.I.); or (G.D.)
| | - Laura Ferrante
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy or (A.M.I.); or (G.L.); or (L.F.); or (E.d.R.); (M.L.); or (A.D.I.); or (G.D.)
| | - Elisabetta de Ruvo
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy or (A.M.I.); or (G.L.); or (L.F.); or (E.d.R.); (M.L.); or (A.D.I.); or (G.D.)
| | - Merigrazia Campanelli
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy or (A.M.I.); or (G.L.); or (L.F.); or (E.d.R.); (M.L.); or (A.D.I.); or (G.D.)
| | - Marialuisa Longo
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy or (A.M.I.); or (G.L.); or (L.F.); or (E.d.R.); (M.L.); or (A.D.I.); or (G.D.)
| | - Andrea Palermo
- College of Medicine and Dentistry, Birmingham B4 6BN, UK;
| | - Alessio Danilo Inchingolo
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy or (A.M.I.); or (G.L.); or (L.F.); or (E.d.R.); (M.L.); or (A.D.I.); or (G.D.)
| | - Gianna Dipalma
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy or (A.M.I.); or (G.L.); or (L.F.); or (E.d.R.); (M.L.); or (A.D.I.); or (G.D.)
| |
Collapse
|
14
|
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
|
15
|
Fukuda R, Pak K, Kiuchi M, Hirata N, Mochimaru N, Tanaka R, Mitsui M, Ohya Y, Yoshida K. Longitudinal Correlations between Molecular Compositions of Stratum Corneum and Breast Milk Factors during Infancy: A Prospective Birth Cohort Study. Nutrients 2024; 16:1897. [PMID: 38931252 PMCID: PMC11206726 DOI: 10.3390/nu16121897] [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: 05/01/2024] [Revised: 05/31/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Breast milk contains numerous factors that are involved in the maturation of the immune system and development of the gut microbiota in infants. These factors include transforming growth factor-β1 and 2, immunoglobin A, and lactoferrin. Breast milk factors may also affect epidermal differentiation and the stratum corneum (SC) barrier in infants, but no studies examining these associations over time during infancy have been reported. In this single-center exploratory study, we measured the molecular components of the SC using confocal Raman spectroscopy at 0, 1, 2, 6, and 12 months of age in 39 infants born at our hospital. Breast milk factor concentrations from their mothers' breast milk were determined. Correlation coefficients for the two datasets were estimated for each molecular component of the SC and breast milk factor at each age and SC depth. The results showed that breast milk factors and molecular components of the SC during infancy were partly correlated with infant age in months and SC depth, suggesting that breast milk factors influence the maturation of the SC components. These findings may improve understanding of the pathogenesis of skin diseases associated with skin barrier abnormalities.
Collapse
Affiliation(s)
- Risa Fukuda
- Division of Dermatology, National Center for Child Health and Development, Tokyo 157-8535, Japan; (R.F.); (R.T.)
| | - Kyongsun Pak
- Division of Biostatistics, Department of Data Management, Center of Clinical Research and Development, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Megumi Kiuchi
- Division of Research and Development, Pigeon Corporation, Ibaraki 300-2495, Japan
| | - Naoko Hirata
- Division of Research and Development, Pigeon Corporation, Ibaraki 300-2495, Japan
| | - Naoko Mochimaru
- Division of Dermatology, National Center for Child Health and Development, Tokyo 157-8535, Japan; (R.F.); (R.T.)
| | - Ryo Tanaka
- Division of Dermatology, National Center for Child Health and Development, Tokyo 157-8535, Japan; (R.F.); (R.T.)
| | - Mari Mitsui
- Center for Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Yukihiro Ohya
- Allergy Center, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Kazue Yoshida
- Division of Dermatology, National Center for Child Health and Development, Tokyo 157-8535, Japan; (R.F.); (R.T.)
- Allergy Center, National Center for Child Health and Development, Tokyo 157-8535, Japan
| |
Collapse
|
16
|
Selma-Royo M, Dubois L, Manara S, Armanini F, Cabrera-Rubio R, Valles-Colomer M, González S, Parra-Llorca A, Escuriet R, Bode L, Martínez-Costa C, Segata N, Collado MC. Birthmode and environment-dependent microbiota transmission dynamics are complemented by breastfeeding during the first year. Cell Host Microbe 2024; 32:996-1010.e4. [PMID: 38870906 PMCID: PMC11183301 DOI: 10.1016/j.chom.2024.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/24/2024] [Accepted: 05/10/2024] [Indexed: 06/15/2024]
Abstract
The composition and maturation of the early-life microbiota are modulated by a number of perinatal factors, whose interplay in relation to microbial vertical transmission remains inadequately elucidated. Using recent strain-tracking methodologies, we analyzed mother-to-infant microbiota transmission in two different birth environments: hospital-born (vaginal/cesarean) and home-born (vaginal) infants and their mothers. While delivery mode primarily explains initial compositional differences, place of birth impacts transmission timing-being early in homebirths and delayed in cesarean deliveries. Transmission patterns vary greatly across species and birth groups, yet certain species, like Bifidobacterium longum, are consistently vertically transmitted regardless of delivery setting. Strain-level analysis of B. longum highlights relevant and consistent subspecies replacement patterns mainly explained by breastfeeding practices, which drive changes in human milk oligosaccharide (HMO) degrading capabilities. Our findings highlight how delivery setting, breastfeeding duration, and other lifestyle preferences collectively shape vertical transmission, impacting infant gut colonization during early life.
Collapse
Affiliation(s)
- Marta Selma-Royo
- Institute of Agrochemistry and Food Technology-Spanish National Research Council (IATA-CSIC), Paterna, Valencia, Spain; Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Léonard Dubois
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Serena Manara
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Federica Armanini
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Raúl Cabrera-Rubio
- Institute of Agrochemistry and Food Technology-Spanish National Research Council (IATA-CSIC), Paterna, Valencia, Spain
| | - Mireia Valles-Colomer
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy; MELIS Department, Universitat Pompeu Fabra, Barcelona, Spain
| | - Sonia González
- Department of Functional Biology, University of Oviedo, Oviedo, Spain; Diet Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Anna Parra-Llorca
- Health Research Institute La Fe, Neonatal Research Group, Division of Neonatology, Valencia, Spain
| | - Ramon Escuriet
- Gerencia de Procesos Integrales de Salud. Area Asistencial, Servicio Catalan de la Salud, Generalitat de Catalunya, Centre for Research in Health and Economics, Universidad Pompeu Fabra, Barcelona, Spain
| | - Lars Bode
- Department of Pediatrics, Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (LRF MOMI CORE), Human Milk Institute (HMI), University of California, San Diego, La Jolla, CA, USA
| | - Cecilia Martínez-Costa
- Department of Pediatrics, Hospital Clínico Universitario, University of Valencia, Spain; Nutrition Research Group of INCLIVA, Valencia, Spain
| | - Nicola Segata
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy.
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology-Spanish National Research Council (IATA-CSIC), Paterna, Valencia, Spain.
| |
Collapse
|
17
|
Sugino KY, Janssen RC, McMahan RH, Zimmerman C, Friedman JE, Jonscher KR. Vertical Transfer of Maternal Gut Microbes to Offspring of Western Diet-Fed Dams Drives Reduced Levels of Tryptophan Metabolites and Postnatal Innate Immune Response. Nutrients 2024; 16:1808. [PMID: 38931163 PMCID: PMC11206590 DOI: 10.3390/nu16121808] [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: 05/09/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Maternal obesity and/or Western diet (WD) is associated with an increased risk of metabolic dysfunction-associated steatotic liver disease (MASLD) in offspring, driven, in part, by the dysregulation of the early life microbiome. Here, using a mouse model of WD-induced maternal obesity, we demonstrate that exposure to a disordered microbiome from WD-fed dams suppressed circulating levels of endogenous ligands of the aryl hydrocarbon receptor (AHR; indole, indole-3-acetate) and TMAO (a product of AHR-mediated transcription), as well as hepatic expression of Il10 (an AHR target), in offspring at 3 weeks of age. This signature was recapitulated by fecal microbial transfer from WD-fed pregnant dams to chow-fed germ-free (GF) lactating dams following parturition and was associated with a reduced abundance of Lactobacillus in GF offspring. Further, the expression of Il10 was downregulated in liver myeloid cells and in LPS-stimulated bone marrow-derived macrophages (BMDM) in adult offspring, suggestive of a hypo-responsive, or tolerant, innate immune response. BMDMs from adult mice lacking AHR in macrophages exhibited a similar tolerogenic response, including diminished expression of Il10. Overall, our study shows that exposure to maternal WD alters microbial metabolites in the offspring that affect AHR signaling, potentially contributing to innate immune hypo-responsiveness and progression of MASLD, highlighting the impact of early life gut dysbiosis on offspring metabolism. Further investigations are warranted to elucidate the complex interplay between maternal diet, gut microbial function, and the development of neonatal innate immune tolerance and potential therapeutic interventions targeting these pathways.
Collapse
Affiliation(s)
- Kameron Y. Sugino
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.Y.S.); (R.C.J.); (J.E.F.)
| | - Rachel C. Janssen
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.Y.S.); (R.C.J.); (J.E.F.)
| | - Rachel H. McMahan
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Chelsea Zimmerman
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Jacob E. Friedman
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.Y.S.); (R.C.J.); (J.E.F.)
- Department of Biochemistry and Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Karen R. Jonscher
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.Y.S.); (R.C.J.); (J.E.F.)
- Department of Biochemistry and Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
18
|
Bornbusch SL, Shinnerl HE, Gentry L, Keady MM, Glick V, Muletz-Wolz CR, Power ML. Local environment shapes milk microbiomes while evolutionary history constrains milk macronutrients in captive cercopithecine primates. Environ Microbiol 2024; 26:e16664. [PMID: 38830671 DOI: 10.1111/1462-2920.16664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 05/16/2024] [Indexed: 06/05/2024]
Abstract
Milk is a complex biochemical fluid that includes macronutrients and microbiota, which, together, are known to facilitate infant growth, mediate the colonization of infant microbiomes, and promote immune development. Examining factors that shape milk microbiomes and milk-nutrient interplay across host taxa is critical to resolving the evolution of the milk environment. Using a comparative approach across four cercopithecine primate species housed at three facilities under similar management conditions, we test for the respective influences of the local environment (housing facility) and host species on milk (a) macronutrients (fat, sugar, and protein), (b) microbiomes (16S rRNA), and (c) predicted microbial functions. We found that milk macronutrients were structured according to host species, while milk microbiomes and predicted function were strongly shaped by the local environment and, to a lesser extent, host species. The milk microbiomes of rhesus macaques (Macaca mulatta) at two different facilities more closely resembled those of heterospecific facility-mates compared to conspecifics at a different facility. We found similar, facility-driven patterns of microbial functions linked to physiology and immune modulation, suggesting that milk microbiomes may influence infant health and development. These results provide novel insight into the complexity of milk and its potential impact on infants across species and environments.
Collapse
Affiliation(s)
- Sally L Bornbusch
- Center for Conservation Genomics, Smithsonian's National Zoo and Conservation Biology Institute, Washington, DC, USA
- Department of Nutrition Science, Smithsonian's National Zoo and Conservation Biology Institute, Washington, DC, USA
| | - Hannah E Shinnerl
- Department of Nutrition Science, Smithsonian's National Zoo and Conservation Biology Institute, Washington, DC, USA
| | - Lindsey Gentry
- Center for Conservation Genomics, Smithsonian's National Zoo and Conservation Biology Institute, Washington, DC, USA
| | - Mia M Keady
- Nelson Institute for Environmental Studies, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Virginia Glick
- Department of Nutrition Science, Smithsonian's National Zoo and Conservation Biology Institute, Washington, DC, USA
- Department of Immunology and Infectious Disease, Harvard University, Harvard School of Public Health, Boston, Massachusetts, USA
| | - Carly R Muletz-Wolz
- Center for Conservation Genomics, Smithsonian's National Zoo and Conservation Biology Institute, Washington, DC, USA
| | - Michael L Power
- Department of Nutrition Science, Smithsonian's National Zoo and Conservation Biology Institute, Washington, DC, USA
- Center for Species Survival, Smithsonian National Zoo and Conservation Biology Institute, Washington, DC, USA
| |
Collapse
|
19
|
Xu R, McLoughlin G, Nicol M, Geddes D, Stinson L. Residents or Tourists: Is the Lactating Mammary Gland Colonized by Residential Microbiota? Microorganisms 2024; 12:1009. [PMID: 38792838 PMCID: PMC11123721 DOI: 10.3390/microorganisms12051009] [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: 04/05/2024] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024] Open
Abstract
The existence of the human milk microbiome has been widely recognized for almost two decades, with many studies examining its composition and relationship to maternal and infant health. However, the richness and viability of the human milk microbiota is surprisingly low. Given that the lactating mammary gland houses a warm and nutrient-rich environment and is in contact with the external environment, it may be expected that the lactating mammary gland would contain a high biomass microbiome. This discrepancy raises the question of whether the bacteria in milk come from true microbial colonization in the mammary gland ("residents") or are merely the result of constant influx from other bacterial sources ("tourists"). By drawing together data from animal, in vitro, and human studies, this review will examine the question of whether the lactating mammary gland is colonized by a residential microbiome.
Collapse
Affiliation(s)
- Ruomei Xu
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia (D.G.)
| | - Grace McLoughlin
- School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia; (G.M.); (M.N.)
| | - Mark Nicol
- School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia; (G.M.); (M.N.)
| | - Donna Geddes
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia (D.G.)
| | - Lisa Stinson
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia (D.G.)
| |
Collapse
|
20
|
Sindi AS, Stinson LF, Gridneva Z, Leghi GE, Netting MJ, Wlodek ME, Muhlhausler BS, Rea A, Trevenen ML, Geddes DT, Payne MS. Maternal dietary intervention during lactation impacts the maternal faecal and human milk microbiota. J Appl Microbiol 2024; 135:lxae024. [PMID: 38323424 DOI: 10.1093/jambio/lxae024] [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: 10/04/2023] [Revised: 12/05/2023] [Accepted: 02/05/2024] [Indexed: 02/08/2024]
Abstract
AIMS To determine the effect of a two-week reduced fat and sugar and increased fibre maternal dietary intervention on the maternal faecal and human milk (HM) microbiomes. METHODS AND RESULTS Faecal swabs and HM samples were collected from mothers (n = 11) immediately pre-intervention, immediately post-intervention, and 4 and 8 weeks post-intervention, and were analysed using full-length 16S rRNA gene sequencing. Maternal macronutrient intake was assessed at baseline and during the intervention. Maternal fat and sugar intake during the intervention were significantly lower than pre-intervention (P = <0.001, 0.005, respectively). Significant changes in the bacterial composition of maternal faeces were detected after the dietary intervention, with decreases in the relative abundance of Bacteroides caccae (P = <0.001) and increases in the relative abundance of Faecalibacillus intestinalis (P = 0.006). In HM, the diet resulted in a significant increase in Cutibacterium acnes (P = 0.001) and a decrease in Haemophilus parainfluenzae (P = <0.001). The effect of the diet continued after the intervention, with faecal swabs and HM samples taken 4 and 8 weeks after the diet showing significant differences compared to baseline. CONCLUSION This pilot study demonstrates that short-term changes in maternal diet during lactation can alter the bacterial composition of the maternal faeces and HM.
Collapse
Affiliation(s)
- Azhar S Sindi
- Division of Obstetrics and Gynaecology, School of Medicine, The University of Western Australia, Subiaco, WA 6008, Australia
- College of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Lisa F Stinson
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia
| | - Zoya Gridneva
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia
| | - Gabriela E Leghi
- School of Agriculture, Food and Wine, The University of Adelaide, Urrbrae, SA 5064, Australia
| | - Merryn J Netting
- Women and Kids Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, SA 5000, Australia
- Discipline of Paediatrics, The University of Adelaide, North Adelaide, SA 5006, Australia
- Women's and Children's Hospital, North Adelaide, SA 5006, Australia
| | - Mary E Wlodek
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Beverly S Muhlhausler
- School of Agriculture, Food and Wine, The University of Adelaide, Urrbrae, SA 5064, Australia
- CSIRO, Adelaide, SA 5000, Australia
| | - Alethea Rea
- Centre for Applied Statistics, The University of Western Australia, Crawley, WA 6009, Australia
- Mathematics and Statistics, Murdoch University, Murdoch, WA 6150, Australia
| | - Michelle L Trevenen
- Centre for Applied Statistics, The University of Western Australia, Crawley, WA 6009, Australia
| | - Donna T Geddes
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia
| | - Matthew S Payne
- Division of Obstetrics and Gynaecology, School of Medicine, The University of Western Australia, Subiaco, WA 6008, Australia
| |
Collapse
|
21
|
Shahi S, Kang T, Fonseka P. Extracellular Vesicles in Pathophysiology: A Prudent Target That Requires Careful Consideration. Cells 2024; 13:754. [PMID: 38727289 PMCID: PMC11083420 DOI: 10.3390/cells13090754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Extracellular vesicles (EVs) are membrane-bound particles released by cells to perform multitudes of biological functions. Owing to their significant implications in diseases, the pathophysiological role of EVs continues to be extensively studied, leading research to neglect the need to explore their role in normal physiology. Despite this, many identified physiological functions of EVs, including, but not limited to, tissue repair, early development and aging, are attributed to their modulatory role in various signaling pathways via intercellular communication. EVs are widely perceived as a potential therapeutic strategy for better prognosis, primarily through utilization as a mode of delivery vehicle. Moreover, disease-associated EVs serve as candidates for the targeted inhibition by pharmacological or genetic means. However, these attempts are often accompanied by major challenges, such as off-target effects, which may result in adverse phenotypes. This renders the clinical efficacy of EVs elusive, indicating that further understanding of the specific role of EVs in physiology may enhance their utility. This review highlights the essential role of EVs in maintaining cellular homeostasis under different physiological settings, and also discusses the various aspects that may potentially hinder the robust utility of EV-based therapeutics.
Collapse
Affiliation(s)
| | | | - Pamali Fonseka
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia; (S.S.); (T.K.)
| |
Collapse
|
22
|
Alba C, Carrera M, Álvarez-Calatayud G, Arroyo R, Fernández L, Rodríguez JM. Evaluation of Safety and Beneficial Health Effects of the Human-Milk Strain Bifidobacterium breve DSM32583: An Infant Pilot Trial. Nutrients 2024; 16:1134. [PMID: 38674825 PMCID: PMC11053739 DOI: 10.3390/nu16081134] [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: 03/15/2024] [Revised: 04/05/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Human milk promotes the growth of bifidobacteria in the infant gut. Adding bifidobacterial species to infant formula may contribute to increasing their presence in the gut of formula-fed infants. Therefore, the safety and anti-infectious effects of Bifidobacterium breve DSM32583, a breast milk isolate, were assessed in a pilot trial involving 3-month-old infants. The infants were randomly assigned to either the probiotic (PG) or the control (CG) groups. All the infants consumed the same formula, although it was supplemented with the strain (1 × 107 cfu/g of formula) in the PG. Overall, 160 infants (80 per group) finished the intervention. Infants in CG gained more weight compared to PG (p < 0.05), but the weights for age Z-scores at 6 months were within the normal distribution for this age group. The rates of infections affecting the gastrointestinal and respiratory tracts and antibiotic therapy were significantly lower in the PG. The bifidobacterial population and the level of short-chain fatty acids were higher (p < 0.05) in the fecal samples of PG infants. No adverse events related to formula consumption were observed. In conclusion, the administration of an infant formula with B. breve DSM32583 was safe and exerted potential beneficial effects on gut health.
Collapse
Affiliation(s)
- Claudio Alba
- Department of Nutrition and Food Science, Complutense University of Madrid, 28040 Madrid, Spain; (C.A.); (R.A.)
| | - Marta Carrera
- Centro de Atención Primaria Silvano, Comunidad de Madrid, 28043 Madrid, Spain;
| | | | - Rebeca Arroyo
- Department of Nutrition and Food Science, Complutense University of Madrid, 28040 Madrid, Spain; (C.A.); (R.A.)
| | - Leónides Fernández
- Department of Galenic Pharmacy and Food Technology, Complutense University of Madrid, 28040 Madrid, Spain;
| | - Juan M. Rodríguez
- Department of Nutrition and Food Science, Complutense University of Madrid, 28040 Madrid, Spain; (C.A.); (R.A.)
| |
Collapse
|
23
|
González A, Fullaondo A, Odriozola A. Impact of evolution on lifestyle in microbiome. ADVANCES IN GENETICS 2024; 111:149-198. [PMID: 38908899 DOI: 10.1016/bs.adgen.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/24/2024]
Abstract
This chapter analyses the interaction between microbiota and humans from an evolutionary point of view. Long-term interactions between gut microbiota and host have been generated as a result of dietary choices through coevolutionary processes, where mutuality of advantage is essential. Likewise, the characteristics of the intestinal environment have made it possible to describe different intrahost evolutionary mechanisms affecting microbiota. For its part, the intestinal microbiota has been of great importance in the evolution of mammals, allowing the diversification of dietary niches, phenotypic plasticity and the selection of host phenotypes. Although the origin of the human intestinal microbial community is still not known with certainty, mother-offspring transmission plays a key role, and it seems that transmissibility between individuals in adulthood also has important implications. Finally, it should be noted that certain aspects inherent to modern lifestyle, including refined diets, antibiotic intake, exposure to air pollutants, microplastics, and stress, could negatively affect the diversity and composition of our gut microbiota. This chapter aims to combine current knowledge to provide a comprehensive view of the interaction between microbiota and humans throughout evolution.
Collapse
Affiliation(s)
- Adriana González
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain.
| | - Asier Fullaondo
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Adrián Odriozola
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain
| |
Collapse
|
24
|
Ajeeb TT, Gonzalez E, Solomons NW, Vossenaar M, Koski KG. Human milk microbiome: associations with maternal diet and infant growth. Front Nutr 2024; 11:1341777. [PMID: 38529196 PMCID: PMC10962684 DOI: 10.3389/fnut.2024.1341777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 02/22/2024] [Indexed: 03/27/2024] Open
Abstract
Introduction Ingestion of human milk (HM) is identified as a significant factor associated with early infant gut microbial colonization, which has been associated with infant health and development. Maternal diet has been associated with the HM microbiome (HMM). However, a few studies have explored the associations among maternal diet, HMM, and infant growth during the first 6 months of lactation. Methods For this cross-sectional study, Mam-Mayan mother-infant dyads (n = 64) were recruited from 8 rural communities in the Western Highlands of Guatemala at two stages of lactation: early (6-46 days postpartum, n = 29) or late (109-184 days postpartum, n = 35). Recruited mothers had vaginally delivered singleton births, had no subclinical mastitis or antibiotic treatments, and breastfed their infants. Data collected at both stages of lactation included two 24-h recalls, milk samples, and infant growth status indicators: head-circumference-for-age-z-score (HCAZ), length-for-age-z-score (LAZ), and weight-for-age-z-score (WAZ). Infants were divided into subgroups: normal weight (WAZ ≥ -1SD) and mildly underweight (WAZ < -1SD), non-stunted (LAZ ≥ -1.5SD) and mildly stunted (LAZ < -1.5SD), and normal head-circumference (HCAZ ≥ -1SD) and smaller head-circumference (HCAZ < -1SD). HMM was identified using 16S rRNA gene sequencing; amplicon analysis was performed with the high-resolution ANCHOR pipeline, and DESeq2 identified the differentially abundant (DA) HMM at the species-level between infant growth groups (FDR < 0.05) in both early and late lactation. Results Using both cluster and univariate analyses, we identified (a) positive correlations between infant growth clusters and maternal dietary clusters, (b) both positive and negative associations among maternal macronutrient and micronutrient intakes with the HMM at the species level and (c) distinct correlations between HMM DA taxa with maternal nutrient intakes and infant z-scores that differed between breast-fed infants experiencing growth faltering and normal growth in early and late lactation. Conclusion Collectively, these findings provide important evidence of the potential influence of maternal diet on the early-life growth of breastfed infants via modulation of the HMM.
Collapse
Affiliation(s)
- Tamara T. Ajeeb
- School of Human Nutrition, McGill University, Montreal, QC, Canada
- Department of Clinical Nutrition, College of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Emmanuel Gonzalez
- Canadian Centre for Computational Genomics, McGill Genome Centre, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada
| | - Noel W. Solomons
- Center for Studies of Sensory Impairment, Aging and Metabolism (CeSSIAM), Guatemala City, Guatemala
| | - Marieke Vossenaar
- Center for Studies of Sensory Impairment, Aging and Metabolism (CeSSIAM), Guatemala City, Guatemala
| | | |
Collapse
|
25
|
Nixarlidou E, Margioula-Siarkou C, Almperis A, Vavoulidis E, Laganà AS, Dinas K, Petousis S. Clinical significance and main parameters promoting the breast‑feeding strategy (Review). MEDICINE INTERNATIONAL 2024; 4:14. [PMID: 38410759 PMCID: PMC10895466 DOI: 10.3892/mi.2024.138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 02/02/2024] [Indexed: 02/28/2024]
Abstract
Breastfeeding provides numerous nutritional and immunological benefits, promotes neurological and cognitive development, and protects against chronic and infectious diseases, rendering it beneficial to the survival and well-being of infants. According to international recommendations, infants should be exclusively breastfed for the first 6 months. However, despite global health recommendations and funding initiatives, exclusive breastfeeding rates remain low worldwide. A number of studies attribute the low rates to factors that can be grouped into demographic, psychosocial, economic and midwifery factors, and outline the profile of each mother who opts to exclusively breastfeed her infant. In addition, the number of previous pregnancies, induced labor, the use of epidurals at birth or the possibility of the newborn being delivered prematurely, and the need for admission to an intensive care unit are the factors that reduce the likelihood of exclusive breastfeeding. Further research is required to understand the factors influencing the initiation and maintenance of exclusive breastfeeding, as international interventions have been ineffective. The aim of the present review was to provide an up-to-date summary of these various factors in an aim to assist health care professionals and policy makers in developing effective interventions with which to promote and support exclusive breastfeeding.
Collapse
Affiliation(s)
- Eleni Nixarlidou
- 2nd Department of Obstetrics and Gynecology, Aristotle University of Thessaloniki, 54624 Thessaloniki, Greece
| | - Chrysoula Margioula-Siarkou
- 2nd Department of Obstetrics and Gynecology, Aristotle University of Thessaloniki, 54624 Thessaloniki, Greece
| | - Aristarchos Almperis
- 2nd Department of Obstetrics and Gynecology, Aristotle University of Thessaloniki, 54624 Thessaloniki, Greece
| | - Eleftherios Vavoulidis
- 2nd Department of Obstetrics and Gynecology, Aristotle University of Thessaloniki, 54624 Thessaloniki, Greece
| | - Antonio Simone Laganà
- Unit of Gynecologic Oncology, ARNAS 'Civico-Di Cristina-Benfratelli', Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, I-90127 Palermo, Italy
| | - Konstantinos Dinas
- 2nd Department of Obstetrics and Gynecology, Aristotle University of Thessaloniki, 54624 Thessaloniki, Greece
| | - Stamatios Petousis
- 2nd Department of Obstetrics and Gynecology, Aristotle University of Thessaloniki, 54624 Thessaloniki, Greece
| |
Collapse
|
26
|
Sharma N, Chaudhary SM, Khungar N, Aulakh SK, Idris H, Singh A, Sharma K. Dietary Influences on Skin Health in Common Dermatological Disorders. Cureus 2024; 16:e55282. [PMID: 38562266 PMCID: PMC10982215 DOI: 10.7759/cureus.55282] [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] [Accepted: 02/29/2024] [Indexed: 04/04/2024] Open
Abstract
The role of diet in the development of skin disorders is well-established, with nutritional deficiency often identified as a risk factor for skin diseases. Imbalances in the skin can be caused by nutritional deficiencies, excessive intake, insufficient nutrients, and hazardous ingredients. Patients frequently inquire about the impact of dietary patterns on skin health when consulting dermatologists in clinical settings. Simultaneously, the popularity of using nutritional supplements containing vitamins, minerals, and nutraceutical blends has been on the rise. It is crucial for dermatologists, primary care physicians, and other healthcare providers to be acquainted with evidence-based dietary interventions, distinguishing them from those that are more market-driven than truly efficacious. This review explores the modification of diet, encompassing both dietary exclusion and supplementation, as a therapeutic approach for conditions such as psoriasis, atopic dermatitis, bullous disease, vitiligo, and alopecia areata. A comprehensive literature search, utilizing the PubMed/Medline, Google Scholar, and Medscape databases, was conducted to investigate the relationship between each nutrient and various inflammatory skin diseases. The findings emphasize the significance of a well-balanced and thoughtfully planned diet in supplying adequate amounts of proteins, vitamins, and minerals to support optimal skin health. Additionally, this comprehensive review navigates through various dietary recommendations, offering insights into their multifaceted impacts on the immune system, gut microbiome, and skin health. The goal is to pave the way for informed and targeted dietary interventions for individuals dealing with food allergies and associated skin conditions.
Collapse
Affiliation(s)
| | - Sachin M Chaudhary
- Internal Medicine, Gujarat Cancer Society (GCS) Medical College, Hospital and Research Centre, Ahmedabad, IND
| | - Niharika Khungar
- Internal Medicine, Sri Guru Ramdas University of Health Science and Research, Amritsar, IND
| | - Smriti K Aulakh
- Internal Medicine, Sri Guru Ramdas University of Health Science and Research, Amritsar, IND
| | - Hadeeqa Idris
- Internal Medicine, Shifa International Hospital, Islamabad, PAK
| | - Ajay Singh
- Internal Medicine, Sri Ram Murti Smarak Institute of Medical Sciences, Bareilly, IND
| | - Kriti Sharma
- Internal Medicine, Government Medical College, Amritsar, Amritsar, IND
| |
Collapse
|
27
|
Qian M, Liu W, Feng X, Yang Z, Liu X, Ma L, Shan Y, Ran N, Yi M, Wei C, Lu C, Wang Y. Alterations in the gut microbiota of toddlers with cow milk protein allergy treated with a partially hydrolyzed formula containing synbiotics: A nonrandomized controlled interventional study. Food Sci Nutr 2024; 12:765-775. [PMID: 38370083 PMCID: PMC10867501 DOI: 10.1002/fsn3.3801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 10/14/2023] [Accepted: 10/18/2023] [Indexed: 02/20/2024] Open
Abstract
Formulas containing intact cow milk protein are appropriate alternatives when human milk (HM) is not feasible. However, for babies with a physician-diagnosed cow milk protein allergy (CMPA), hydrolyzed formulas are needed. We conducted a 3-month, open-label, nonrandomized concurrent controlled trial (ChiCTR2100046909) between June 2021 and October 2022 in Qingdao City, China. In this study, CMPA toddlers were fed with a partially hydrolyzed formula containing synbiotics (pHF, n = 43) and compared with healthy toddlers fed a regular intact protein formula (IF, n = 45) or HM (n = 21). The primary endpoint was weight gain; the secondary endpoints were changes in body length and head circumference of both CMPA and healthy toddlers after 3-month feeding; and the exploratory outcomes were changes in gut microbiota composition. After 3 months, there were no significant group differences for length-for-age, weight-for-age, or head circumference-for-age Z scores. In the gut microbiota, pHF feeding increased its richness and diversity, similar to those of IF-fed and HM-fed healthy toddlers. Compared with healthy toddlers, the toddlers with CMPA showed an increased abundance of phylum Bacteroidota, Firmicutes, class Clostridia, and Bacteroidia, and a decreased abundance of class Negativicutes, while pHF feeding partly eliminated these original differences. Moreover, pHF feeding increased the abundance of short-chain fatty acid producers. Our data suggested that this pHF partly simulated the beneficial effects of HM and shifted the gut microbiota of toddlers with CMPA toward that of healthy individuals. In conclusion, this synbiotic-containing pHF might be an appropriate alternative for toddlers with CMPA.
Collapse
Affiliation(s)
- Mengyao Qian
- The Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Wei Liu
- The Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Xueying Feng
- The Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Zhaochuan Yang
- The Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Xiaomei Liu
- The Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Liang Ma
- The Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Yanchun Shan
- The Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Ni Ran
- The Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Mingji Yi
- The Affiliated Hospital of Qingdao UniversityQingdaoChina
| | | | - Chenyang Lu
- School of Marine ScienceNingbo UniversityNingboChina
| | - Yanxia Wang
- The Affiliated Hospital of Qingdao UniversityQingdaoChina
| |
Collapse
|
28
|
Wong PY, Yip C, Lemberg DA, Day AS, Leach ST. Evolution of a Pathogenic Microbiome. J Clin Med 2023; 12:7184. [PMID: 38002796 PMCID: PMC10672640 DOI: 10.3390/jcm12227184] [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: 09/22/2023] [Revised: 11/16/2023] [Accepted: 11/16/2023] [Indexed: 11/26/2023] Open
Abstract
The process of microbiome development arguably begins before birth. Vertical transmission of bacteria from the mother to the infant is a keystone event in microbiome development. Subsequent to birth, the developing microbiome is vulnerable to influence from a wide range of factors. Additionally, the microbiome can influence the health and development of the host infant. This intricate interaction of the gastrointestinal microbiome and the host has been described as both symbiotic and dysbiotic. Defining these terms, a symbiotic microbiome is where the microbiome and host provide mutual benefit to each other. A pathogenic microbiome, or more precisely a gastrointestinal microbiome associated with disease, is increasing described as dysbiotic. This review seeks to investigate the factors that contribute to evolving a disease-causing or 'dysbiotic' microbiome. This review covers the development of the gastrointestinal microbiome in infants, the interaction of the microbiome with the host, and its contribution to host immunity and investigates specific features of the gastrointestinal microbiome that are associated with disease.
Collapse
Affiliation(s)
- Pui Yin Wong
- Discipline of Paediatrics, School of Clinical Medicine, University of NSW, Sydney 2052, Australia; (P.Y.W.); (C.Y.); (A.S.D.)
| | - Carmen Yip
- Discipline of Paediatrics, School of Clinical Medicine, University of NSW, Sydney 2052, Australia; (P.Y.W.); (C.Y.); (A.S.D.)
| | - Daniel A. Lemberg
- Department of Gastroenterology, Sydney Children’s Hospital Randwick, Sydney 2031, Australia;
| | - Andrew S. Day
- Discipline of Paediatrics, School of Clinical Medicine, University of NSW, Sydney 2052, Australia; (P.Y.W.); (C.Y.); (A.S.D.)
- Department of Gastroenterology, Sydney Children’s Hospital Randwick, Sydney 2031, Australia;
- Department of Paediatrics, University of Otago, Christchurch 8011, New Zealand
| | - Steven T. Leach
- Discipline of Paediatrics, School of Clinical Medicine, University of NSW, Sydney 2052, Australia; (P.Y.W.); (C.Y.); (A.S.D.)
| |
Collapse
|
29
|
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
|
30
|
Myckatyn TM, Duran Ramirez JM, Walker JN, Hanson BM. Management of Biofilm with Breast Implant Surgery. Plast Reconstr Surg 2023; 152:919e-942e. [PMID: 37871028 DOI: 10.1097/prs.0000000000010791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
LEARNING OBJECTIVES After studying this article, the participant should be able to: 1. Understand how bacteria negatively impact aesthetic and reconstructive breast implants. 2. Understand how bacteria infect breast implants. 3. Understand the evidence associated with common implant infection-prevention strategies, and their limitations. 4. Understand why implementation of bacteria-mitigation strategies such as antibiotic administration or "no-touch" techniques may not indefinitely prevent breast implant infection. SUMMARY Bacterial infection of aesthetic and reconstructive breast implants is a common and expensive problem. Subacute infections or chronic capsular contractures leading to device explantation are the most commonly documented sequelae. Although bench and translational research underscores the complexities of implant-associated infection, high-quality studies with adequate power, control groups, and duration of follow-up are lacking. Common strategies to minimize infections use antibiotics-administered systemically, in the breast implant pocket, or by directly bathing the implant before insertion-to limit bacterial contamination. Limiting contact between the implant and skin or breast parenchyma represents an additional common strategy. The clinical prevention of breast implant infection is challenged by the clean-contaminated nature of breast parenchyma, and the variable behavior of not only specific bacterial species but also their strains. These factors impact bacterial virulence and antibiotic resistance.
Collapse
Affiliation(s)
- Terence M Myckatyn
- From the Division of Plastic and Reconstructive Surgery, Washington University in St. Louis School of Medicine
| | | | - Jennifer N Walker
- Department of Microbiology and Molecular Genetics
- Center for Infectious Diseases, Department of Epidemiology, School of Public Health, University of Texas Health Science Center at Houston
| | - Blake M Hanson
- Center for Antimicrobial Resistance and Microbial Genomics, McGovern Medical School
- Center for Infectious Diseases, Department of Epidemiology, School of Public Health, University of Texas Health Science Center at Houston
| |
Collapse
|
31
|
Zhao H, Ma X, Song J, Jiang J, Fei X, Luo Y, Ru Y, Luo Y, Gao C, Kuai L, Li B. From gut to skin: exploring the potential of natural products targeting microorganisms for atopic dermatitis treatment. Food Funct 2023; 14:7825-7852. [PMID: 37599562 DOI: 10.1039/d3fo02455e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Atopic dermatitis (AD) is the most common chronic inflammatory skin disease. Recent studies have revealed that interactions between pathogenic microorganisms, which have a tendency to parasitize the skin of AD patients, play a significant role in the progression of the disease. Furthermore, specific species of commensal bacteria in the human intestinal tract can have a profound impact on the immune system by promoting inflammation and pruritogenesis in AD, while also regulating adaptive immunity. Natural products (NPs) have emerged as promising agents for the treatment of various diseases. Consequently, there is growing interest in utilizing natural products as a novel therapeutic approach for managing AD, with a focus on modulating both skin and gut microbiota. In this review, we discuss the mechanisms and interplay between the skin and gut microbiota in relation to AD. Additionally, we provide a comprehensive overview of recent clinical and fundamental research on NPs targeting the skin and gut microbiota for AD treatment. We anticipate that our work will contribute to the future development of NPs and facilitate research on microbial mechanisms, based on the efficacy of NPs in treating AD.
Collapse
Affiliation(s)
- Hang Zhao
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xin Ma
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Jiankun Song
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Jingsi Jiang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Xiaoya Fei
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Yue Luo
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Yi Ru
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ying Luo
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chunjie Gao
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Le Kuai
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Bin Li
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
32
|
Dos Santos SJ, Shukla I, Hill JE, Money DM. Birth Mode Does Not Determine the Presence of Shared Bacterial Strains between the Maternal Vaginal Microbiome and the Infant Stool Microbiome. Microbiol Spectr 2023; 11:e0061423. [PMID: 37338388 PMCID: PMC10433807 DOI: 10.1128/spectrum.00614-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 06/04/2023] [Indexed: 06/21/2023] Open
Abstract
Dysbiosis of the neonatal gut microbiome during early life has been suggested as the missing link that may explain higher rates of certain diseases in caesarean section-delivered infants. Many studies report delivery mode-related dysbiosis in infants due to a lack of maternal vaginal microbiome exposure, prompting interventions to correct the neonatal gut microbiome by transferring these missing microbes after caesarean delivery. The maternal vaginal microbiome is among the first microbial exposures that many infants experience, yet little is known about the extent of direct transmission of maternal vaginal microbes. As part of the Maternal Microbiome Legacy Project, we aimed to determine if maternal vaginal bacteria are vertically transmitted to infants. We employed cpn60 microbiome profiling, culture-based screening, molecular strain typing, and whole-genome sequencing to determine whether identical maternal vaginal strains were present in infant stool microbiomes. We identified identical cpn60 sequence variants in both halves of maternal-infant dyads in 204 of 585 Canadian women and their newborn infants (38.9%). The same species of Bifidobacterium and Enterococcus were cultured from maternal and corresponding infant samples in 33 and 13 of these mother-infant dyads, respectively. Pulsed-field gel electrophoresis and whole-genome sequencing determined that near-identical strains were detected in these dyads irrespective of delivery mode, indicating an alternative source in cases of caesarean delivery. Overall, we demonstrated that vertical transmission of maternal vaginal microbiota is likely limited and that transmission from other maternal body sites, such as the gut and breast milk, may compensate for the lack of maternal vaginal microbiome exposure during caesarean delivery. IMPORTANCE The importance of the gut microbiome in human health and disease is widely recognized, and there has been a growing appreciation that alterations in gut microbiome composition during a "critical window" of development may impact health in later life. Attempts to correct gut microbiome dysbiosis related to birth mode are underpinned by the assumption that the lack of exposure to maternal vaginal microbes during caesarean delivery is responsible for dysbiosis. Here, we demonstrate that there is limited transmission of the maternal vaginal microbiome to the neonatal gut, even in cases of vaginal delivery. Furthermore, the presence of identical strains shared between mothers and infants in early life, even in cases of caesarean delivery, highlights compensatory microbial exposures and sources for the neonatal stool microbiome other than the maternal vagina.
Collapse
Affiliation(s)
- Scott J. Dos Santos
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Ishika Shukla
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Janet E. Hill
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Deborah M. Money
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Microbiology and Immunology, Faculty of Science, University of British Columbia, Vancouver, British Columbia, Canada
- Women’s Health Research Institute, B.C. Women’s Hospital, Vancouver, British Columbia, Canada
| | - The Maternal Microbiome Legacy Project Team
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Microbiology and Immunology, Faculty of Science, University of British Columbia, Vancouver, British Columbia, Canada
- Women’s Health Research Institute, B.C. Women’s Hospital, Vancouver, British Columbia, Canada
| |
Collapse
|
33
|
Boopathi S, Priya PS, Haridevamuthu B, Nayak SPRR, Chandrasekar M, Arockiaraj J, Jia AQ. Expanding germ-organ theory: Understanding non-communicable diseases through enterobacterial translocation. Pharmacol Res 2023; 194:106856. [PMID: 37460001 DOI: 10.1016/j.phrs.2023.106856] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/02/2023] [Accepted: 07/14/2023] [Indexed: 07/29/2023]
Abstract
Diverse microbial communities colonize different habitats of the human body, including gut, oral cavity, nasal cavity and tissues. These microbial communities are known as human microbiome, plays a vital role in maintaining the health. However, changes in the composition and functions of human microbiome can result in chronic low-grade inflammation, which can damage the epithelial cells and allows pathogens and their toxic metabolites to translocate into other organs such as the liver, heart, and kidneys, causing metabolic inflammation. This dysbiosis of human microbiome has been directly linked to the onset of several non-communicable diseases. Recent metabolomics studies have revealed that pathogens produce several uraemic toxins. These metabolites can serve as inter-kingdom signals, entering the circulatory system and altering host metabolism, thereby aggravating a variety of diseases. Interestingly, Enterobacteriaceae, a critical member of Proteobacteria, has been commonly associated with several non-communicable diseases, and the abundance of this family has been positively correlated with uraemic toxin production. Hence, this review provides a comprehensive overview of Enterobacterial translocation and their metabolites role in non-communicable diseases. This understanding may lead to the identification of novel biomarkers for each metabolic disease as well as the development of novel therapeutic drugs.
Collapse
Affiliation(s)
- Seenivasan Boopathi
- Hainan General Hospital, Hainan affiliated hospital of Hainan Medical University, Haikou 570311, China; Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603 203, Chengalpattu District, Tamil Nadu, India
| | - P Snega Priya
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603 203, Chengalpattu District, Tamil Nadu, India
| | - B Haridevamuthu
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603 203, Chengalpattu District, Tamil Nadu, India
| | - S P Ramya Ranjan Nayak
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603 203, Chengalpattu District, Tamil Nadu, India
| | - Munisamy Chandrasekar
- Department of Veterinary Clinical Medicine, Madras Veterinary College, Chennai, Tamil Nadu, India
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603 203, Chengalpattu District, Tamil Nadu, India.
| | - Ai-Qun Jia
- Hainan General Hospital, Hainan affiliated hospital of Hainan Medical University, Haikou 570311, China.
| |
Collapse
|
34
|
Kolasinac SS, Moe L, Rootwelt V, Sørum H. Bacteria in Normal Canine Milk Analyzed by Blood Agar Medium. Animals (Basel) 2023; 13:2206. [PMID: 37444004 DOI: 10.3390/ani13132206] [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: 06/02/2023] [Revised: 06/27/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Studies of microbiota in normal canine milk from healthy dams are sparse. As is the case with blood and urine, it was considered that milk contains no microbiota. Any discovery of bacteria in canine milk is, therefore, often noted to be a result of contamination during sampling or interpreted as mastitis and treated with antibiotics. Milk was collected twice within 19 days after natural parturition from 11 lactating dams, with no general or local clinical signs of mastitis or other disease. The skin and teats were prepared with an antimicrobial protocol prior to each milk sampling. In total, 210 milk samples were collected and assessed for a number of bacterial colonies grown on each plate. Bacterial growth was detected in 180 samples (86%). Staphylococcus pseudintermedius, Enterococcus spp., Clostridium spp., Coagulase-Negative Staphylococci (CoNS), Streptococcus spp., Streptococcus canis, Bacillus spp., Pasteurella spp., and Escherichia coli were identified from pure and/or mixed bacterial growth, listed in descending order of occurrence. Despite the small sample size, the consistent occurrence of bacteria in early postpartum dams indicates a genuine occurrence of bacteria in canine milk, rather than random contamination. The finding of bacteria in the milk of dams should not, therefore, be the sole argument for the diagnosis of mastitis.
Collapse
Affiliation(s)
- Sabina Sibcic Kolasinac
- Department of Companion Animal Clinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Elizabeth Stephansens vei 15, 1433 Ås, Norway
| | - Lars Moe
- Department of Companion Animal Clinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Elizabeth Stephansens vei 15, 1433 Ås, Norway
| | - Vibeke Rootwelt
- Department of Companion Animal Clinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Elizabeth Stephansens vei 15, 1433 Ås, Norway
| | - Henning Sørum
- Department of Paraclinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Elizabeth Stephansens vei 15, 1433 Ås, Norway
| |
Collapse
|
35
|
Qian X, Tian P, Lin G, Xu X, Wang G, Zhang H, Chen W. Detection of colonization capacity of probiotic Bifidobacterium breve CCFM1025 in the human gut. Future Microbiol 2023; 18:595-606. [PMID: 37314317 DOI: 10.2217/fmb-2022-0131] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023] Open
Abstract
Aim: To detect the gut colonization capacity of Bifidobacterium breve CCFM1025 with clinical antidepressant-like effects. Materials & methods: A unique gene sequence of B. breve CCFM1025 was discovered based on the genome analysis of 104 B. breve strains and a strain-specific primer (1025T5) was designed. In vitro and in vivo samples were used to validate the specificity and quantitative capability of this primer in the PCR system. Results: Quantitative PCR using strain-specific primers enabled absolute quantification of CCFM1025 in fecal samples within 104-1010 cells/g (R2 >0.99). CCFM1025 remained highly detectable in volunteer feces 14 days after cessation of administration, demonstrating its favorable colonization characteristics. Conclusion: CCFM1025 can colonize the healthy human gut.
Collapse
Affiliation(s)
- Xin Qian
- State Key Laboratory of Food Science & Resources, 1800 Lihu Avenue, Wuxi, Jiangsu, 214122, China
- School of Food Science & Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Peijun Tian
- State Key Laboratory of Food Science & Resources, 1800 Lihu Avenue, Wuxi, Jiangsu, 214122, China
- School of Food Science & Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Guopeng Lin
- State Key Laboratory of Food Science & Resources, 1800 Lihu Avenue, Wuxi, Jiangsu, 214122, China
- School of Food Science & Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Xinglong Xu
- Jingjiang Chinese Medicine Hospital, Taizhou, Jiangsu, 214500, China
| | - Gang Wang
- State Key Laboratory of Food Science & Resources, 1800 Lihu Avenue, Wuxi, Jiangsu, 214122, China
- School of Food Science & Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, 214122, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, 225004, China
| | - Hao Zhang
- State Key Laboratory of Food Science & Resources, 1800 Lihu Avenue, Wuxi, Jiangsu, 214122, China
- School of Food Science & Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, 214122, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, 225004, China
| | - Wei Chen
- State Key Laboratory of Food Science & Resources, 1800 Lihu Avenue, Wuxi, Jiangsu, 214122, China
- School of Food Science & Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, 214122, China
| |
Collapse
|
36
|
Golubkova A, Hunter CJ. Development of the Neonatal Intestinal Barrier, Microbiome, and Susceptibility to NEC. Microorganisms 2023; 11:1247. [PMID: 37317221 PMCID: PMC10221463 DOI: 10.3390/microorganisms11051247] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/28/2023] [Accepted: 05/04/2023] [Indexed: 06/16/2023] Open
Abstract
The function of the intestinal barrier is partially dependent on host maturity and the colonization patterns of the microbiome to which it is exposed. Premature birth and stressors of neonatal intensive care unit (NICU)-related support (e.g., antibiotics, steroids, etc.) can alter the host internal environment resulting in changes in the intestinal barrier. Pathogenic microbial proliferation and breach of the immature intestinal barrier are proposed to be crucial steps in the development of neonatal diseases such as necrotizing enterocolitis. This article will review the current literature on the intestinal barrier in the neonatal gut, the consequences of microbiome development for this defense system, and how prematurity can influence neonatal susceptibility to gastrointestinal infection.
Collapse
Affiliation(s)
| | - Catherine J. Hunter
- Division of Pediatric Surgery, Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
37
|
Melekoglu E, Yılmaz B, Çevik A, Gökyıldız Sürücü Ş, Avcıbay Vurgeç B, Gözüyeşil E, Sharma H, Boyan N, Ozogul F. The Impact of the Human Milk Microbiota in the Prevention of Disease and Infant Health. Breastfeed Med 2023. [PMID: 37140562 DOI: 10.1089/bfm.2022.0292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Background: Human milk is recognized as an ideal food for newborns and infants owing to the presence of various nutritive factors, including healthy bacteria. Aim/Objective: This review aimed to understand the effects of human milk microbiota in both the prevention of disease and the health of infants. Methods: Data were obtained from PubMed, Scopus, Web of Science, clinical trial registries, Dergipark, and Türk Atıf Dizini up to February 2023 without language restrictions. Results: It is considered that the first human milk microbiota ingested by the newborn creates the initial microbiome of the gut system, which in turn influences the development and maturation of immunity. Bacteria present in human milk modulate the anti-inflammatory response by releasing certain cytokines, protecting the newborn against certain infections. Therefore, certain bacterial strains isolated from human milk could serve as potential probiotics for various therapeutic applications. Conclusions: In this review, the origin and significance of human milk bacteria have been highlighted along with certain factors influencing the composition of human milk microbiota. In addition, it also summarizes the health benefits of human milk as a protective agent against certain diseases and ailments.
Collapse
Affiliation(s)
- Ebru Melekoglu
- Department of Nutrition and Dietetics, Cukurova University, Adana, Turkey
| | - Birsen Yılmaz
- Department of Nutrition and Dietetics, Cukurova University, Adana, Turkey
| | - Ayseren Çevik
- Department of Midwifery, Cukurova University, Adana, Turkey
| | | | | | - Ebru Gözüyeşil
- Department of Midwifery, Cukurova University, Adana, Turkey
| | - Heena Sharma
- Food Technology Lab, Dairy Technology Division, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Neslihan Boyan
- Department of Anatomy, Faculty of Medicine, Cukurova University, Adana, Turkey
| | - Fatih Ozogul
- Department of Seafood Processing Technology, Faculty of Fisheries, Cukurova University, Adana, Turkey
| |
Collapse
|
38
|
Oddi S, Mantziari A, Huber P, Binetti A, Salminen S, Collado MC, Vinderola G. Human Milk Microbiota Profile Affected by Prematurity in Argentinian Lactating Women. Microorganisms 2023; 11:microorganisms11041090. [PMID: 37110513 PMCID: PMC10145235 DOI: 10.3390/microorganisms11041090] [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: 02/17/2023] [Revised: 03/10/2023] [Accepted: 03/30/2023] [Indexed: 04/29/2023] Open
Abstract
To study (16S rRNA-sequencing) the impact of gestational and corrected ages on the microbiota profile of human milk (HM) of mothers that delivered full-term and pre-term children, HM samples were obtained and classified according to the gestational age as group T (full-term births ≥37 weeks), and group P (pre-term births <37 weeks). Group P was longitudinally followed, and the samples were collected at the full-term corrected gestational age: when the chronological age plus the gestational age were ≥37 weeks (PT group). The HM microbiota composition differed depending on the gestational age (T vs. P). Group T had lower levels of Staphylococcus and higher levels of Rothia and Streptococcus, as compared to group P. The alpha Simpson diversity value was higher in group T than in P, whereas no differences were found between groups T and PT, suggesting a microbial evolution of the composition of group P towards group T over chronological age. Full-term delivery was associated with a greater diversity of microbes in HM. The microbial composition of pre-term HM, at the corrected age, did not show significant differences, as compared to the samples obtained from the full-term group, suggesting that it would be appropriate to consider the corrected age in terms of the composition and the diversity of the milk in future studies.
Collapse
Affiliation(s)
- Sofía Oddi
- Instituto de Lactología Industrial (INLAIN, UNL-CONICET), Facultad de Ingeniería Química, Universidad Nacional del Litoral, Santa Fe 3000, Argentina
| | - Anastasia Mantziari
- Functional Foods Forum, Faculty of Medicine, University of Turku, 20520 Turku, Finland
| | - Paula Huber
- Laboratorio de Plancton, Instituto Nacional de Limnología (INALI, UNL-CONICET), Universidad Nacional del Litoral, Santa Fe 3000, Argentina
- Departamento de Hydrobiologia, Universidade Federal de São Carlos (UFSCar), Rodovia Washington Luiz, São Carlos 13565-905, SP, Brazil
| | - Ana Binetti
- Instituto de Lactología Industrial (INLAIN, UNL-CONICET), Facultad de Ingeniería Química, Universidad Nacional del Litoral, Santa Fe 3000, Argentina
| | - Seppo Salminen
- Functional Foods Forum, Faculty of Medicine, University of Turku, 20520 Turku, Finland
| | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), 46980 Valencia, Spain
| | - Gabriel Vinderola
- Instituto de Lactología Industrial (INLAIN, UNL-CONICET), Facultad de Ingeniería Química, Universidad Nacional del Litoral, Santa Fe 3000, Argentina
| |
Collapse
|
39
|
Couvillion SP, Mostoller KE, Williams JE, Pace RM, Stohel IL, Peterson HK, Nicora CD, Nakayasu ES, Webb-Robertson BJM, McGuire MA, McGuire MK, Metz TO. Interrogating the role of the milk microbiome in mastitis in the multi-omics era. Front Microbiol 2023; 14:1105675. [PMID: 36819069 PMCID: PMC9932517 DOI: 10.3389/fmicb.2023.1105675] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
There is growing interest in a functional understanding of milk-associated microbiota as there is ample evidence that host-associated microbial communities play an active role in host health and phenotype. Mastitis, characterized by painful inflammation of the mammary gland, is prevalent among lactating humans and agricultural animals and is associated with significant clinical and economic consequences. The etiology of mastitis is complex and polymicrobial and correlative studies have indicated alterations in milk microbial community composition. Recent evidence is beginning to suggest that a causal relationship may exist between the milk microbiota and host phenotype in mastitis. Multi-omic approaches can be leveraged to gain a mechanistic, molecular level understanding of how the milk microbiome might modulate host physiology, thereby informing strategies to prevent and ameliorate mastitis. In this paper, we review existing studies that have utilized omics approaches to investigate the role of the milk microbiome in mastitis. We also summarize the strengths and challenges associated with the different omics techniques including metagenomics, metatranscriptomics, metaproteomics, metabolomics and lipidomics and provide perspective on the integration of multiple omics technologies for a better functional understanding of the milk microbiome.
Collapse
Affiliation(s)
- Sneha P. Couvillion
- Pacific Northwest National Laboratory, Earth and Biological Sciences Directorate, Richland, WA, United States,*Correspondence: Sneha P. Couvillion, ✉
| | - Katie E. Mostoller
- Pacific Northwest National Laboratory, Earth and Biological Sciences Directorate, Richland, WA, United States
| | - Janet E. Williams
- Department of Animal, Veterinary, and Food Sciences, University of Idaho, Moscow, ID, United States
| | - Ryan M. Pace
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID, United States
| | - Izabel L. Stohel
- Pacific Northwest National Laboratory, Earth and Biological Sciences Directorate, Richland, WA, United States
| | - Haley K. Peterson
- Department of Animal, Veterinary, and Food Sciences, University of Idaho, Moscow, ID, United States
| | - Carrie D. Nicora
- Pacific Northwest National Laboratory, Earth and Biological Sciences Directorate, Richland, WA, United States
| | - Ernesto S. Nakayasu
- Pacific Northwest National Laboratory, Earth and Biological Sciences Directorate, Richland, WA, United States
| | - Bobbie-Jo M. Webb-Robertson
- Pacific Northwest National Laboratory, Earth and Biological Sciences Directorate, Richland, WA, United States
| | - Mark A. McGuire
- Department of Animal, Veterinary, and Food Sciences, University of Idaho, Moscow, ID, United States
| | - Michelle K. McGuire
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID, United States
| | - Thomas O. Metz
- Pacific Northwest National Laboratory, Earth and Biological Sciences Directorate, Richland, WA, United States,Thomas O. Metz, ✉
| |
Collapse
|
40
|
Leech SM, Gilbert MC, Clifton VL, Kumar S, Rae KM, Borg D, Dekker Nitert M. Insufficient Evidence of a Breastmilk Microbiota at Six-Weeks Postpartum: A Pilot Study. Nutrients 2023; 15:nu15030696. [PMID: 36771402 PMCID: PMC9919471 DOI: 10.3390/nu15030696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/25/2023] [Accepted: 01/28/2023] [Indexed: 01/31/2023] Open
Abstract
Breastmilk is thought to influence the infant gut by supplying prebiotics in the form of human milk oligosaccharides and potentially seeding the gut with breastmilk microbes. However, the presence of a breastmilk microbiota and origins of these microbes are still debated. As a pilot study, we assessed the microbes present in expressed breastmilk at six-weeks postpartum using shotgun metagenomic sequencing in a heterogenous cohort of women who delivered by vaginal (n = 8) and caesarean delivery (n = 8). In addition, we estimated the microbial load of breastmilk at six-weeks post-partum with quantitative PCR targeting the 16S rRNA gene. Breastmilk at six-weeks postpartum had a low microbial mass, comparable with PCR no-template and extraction controls. Microbes identified through metagenomic sequencing were largely consistent with skin and oral microbes, with four samples returning no identifiable bacterial sequences. Our results do not provide convincing evidence for the existence of a breastmilk microbiota at six-weeks postpartum. It is more likely that microbes present in breastmilk are sourced by ejection from the infant's mouth and from surrounding skin, as well as contamination during sampling and processing.
Collapse
Affiliation(s)
- Sophie M. Leech
- School of Chemistry and Molecular Biosciences, The University of Queensland, Saint Lucia, QLD 4072, Australia
| | - Morgan C. Gilbert
- School of Chemistry and Molecular Biosciences, The University of Queensland, Saint Lucia, QLD 4072, Australia
| | - Vicki L. Clifton
- Pregnancy and Development Group, Mater Research Institute, South Brisbane, QLD 4101, Australia
- Faculty of Medicine, The University of Queensland, Saint Lucia, QLD 4072, Australia
| | - Sailesh Kumar
- Faculty of Medicine, The University of Queensland, Saint Lucia, QLD 4072, Australia
- Centre for Maternal and Fetal Medicine, Mater Mothers’ Hospital, Brisbane, QLD 4101, Australia
| | - Kym M. Rae
- Faculty of Medicine, The University of Queensland, Saint Lucia, QLD 4072, Australia
- Indigenous Health, Mater Research Institute, South Brisbane, QLD 4101, Australia
| | - Danielle Borg
- Pregnancy and Development Group, Mater Research Institute, South Brisbane, QLD 4101, Australia
- Faculty of Medicine, The University of Queensland, Saint Lucia, QLD 4072, Australia
| | - Marloes Dekker Nitert
- School of Chemistry and Molecular Biosciences, The University of Queensland, Saint Lucia, QLD 4072, Australia
- Correspondence:
| |
Collapse
|
41
|
Mills S, Yang B, Smith GJ, Stanton C, Ross RP. Efficacy of Bifidobacterium longum alone or in multi-strain probiotic formulations during early life and beyond. Gut Microbes 2023; 15:2186098. [PMID: 36896934 PMCID: PMC10012958 DOI: 10.1080/19490976.2023.2186098] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 02/24/2023] [Indexed: 03/11/2023] Open
Abstract
The significance of Bifidobacterium to human health can be appreciated from its early colonization of the neonatal gut, where Bifidobacterium longum represents the most abundant species. While its relative abundance declines with age, it is further reduced in several diseases. Research into the beneficial properties of B. longum has unveiled a range of mechanisms, including the production of bioactive molecules, such as short-chain fatty acids, polysaccharides, and serine protease inhibitors. From its intestinal niche, B. longum can have far-reaching effects in the body influencing immune responses in the lungs and even skin, as well as influencing brain activity. In this review, we present the biological and clinical impacts of this species on a range of human conditions beginning in neonatal life and beyond. The available scientific evidence reveals a strong rationale for continued research and further clinical trials that investigate the ability of B. longum to treat or prevent a range of diseases across the human lifespan.
Collapse
Affiliation(s)
- Susan Mills
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Bo Yang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | | | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Food Biosciences Department, Teagasc Food Research Centre, Co Cork, Ireland
| | - R. Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
42
|
Tian M, Li Q, Zheng T, Yang S, Chen F, Guan W, Zhang S. Maternal microbe-specific modulation of the offspring microbiome and development during pregnancy and lactation. Gut Microbes 2023; 15:2206505. [PMID: 37184203 DOI: 10.1080/19490976.2023.2206505] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/16/2023] Open
Abstract
The maternal microbiome is essential for the healthy growth and development of offspring and has long-term effects later in life. Recent advances indicate that the maternal microbiome begins to regulate fetal health and development during pregnancy. Furthermore, the maternal microbiome continues to affect early microbial colonization via birth and breastfeeding. Compelling evidence indicates that the maternal microbiome is involved in the regulation of immune and brain development and affects the risk of related diseases. Modulating offspring development by maternal diet and probiotic intervention during pregnancy and breastfeeding could be a promising therapy in the future. In this review, we summarize and discuss the current understanding of maternal microbiota development, perinatal microbial metabolite transfer, mother-to-infant microbial transmission during/after birth and its association with immune and brain development as well as corresponding diseases.
Collapse
Affiliation(s)
- Min Tian
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qihui Li
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Tenghui Zheng
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Siwang Yang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Fang Chen
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
| | - Wutai Guan
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
| | - Shihai Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
43
|
Lundgren SN, Madan JC, Karagas MR, Morrison HG, Christensen BC, Hoen AG. Human milk-associated bacterial communities associate with the infant gut microbiome over the first year of life. Front Microbiol 2023; 14:1164553. [PMID: 37138613 PMCID: PMC10149717 DOI: 10.3389/fmicb.2023.1164553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 03/27/2023] [Indexed: 05/05/2023] Open
Abstract
Introduction Microbial communities inhabiting the human infant gut are important for immune system development and lifelong health. One critical exposure affecting the bacterial colonization of the infant gut is consumption of human milk, which contains diverse microbial communities and prebiotics. We hypothesized that human milk-associated microbial profiles are associated with those of the infant gut. Methods Maternal-infant dyads enrolled in the New Hampshire Birth Cohort Study (n = 189 dyads) contributed breast milk and infant stool samples collected approximately at 6 weeks, 4 months, 6 months, 9 months, and 12 months postpartum (n = 572 samples). Microbial DNA was extracted from milk and stool and the V4-V5 region of the bacterial 16S rRNA gene was sequenced. Results Clustering analysis identified three breast milk microbiome types (BMTs), characterized by differences in Streptococcus, Staphylococcus, Pseudomonas, Acinetobacter, and microbial diversity. Four 6-week infant gut microbiome types (6wIGMTs) were identified, differing in abundances of Bifidobacterium, Bacteroides, Clostridium, Streptococcus, and Escherichia/Shigella, while two 12-month IGMTs (12mIGMTs) differed primarily by Bacteroides presence. At 6 weeks, BMT was associated with 6wIGMT (Fisher's exact test value of p = 0.039); this association was strongest among infants delivered by Cesarean section (Fisher's exact test value of p = 0.0028). The strongest correlations between overall breast milk and infant stool microbial community structures were observed when comparing breast milk samples to infant stool samples collected at a subsequent time point, e.g., the 6-week breast milk microbiome associated with the 6-month infant gut microbiome (Mantel test Z-statistic = 0.53, value of p = 0.001). Streptoccous and Veillonella species abundance were correlated in 6-week milk and infant stool, and 4- and 6-month milk Pantoea species were associated with infant stool Lachnospiraceae genera at 9 and 12 months. Discussion We identified clusters of human milk and infant stool microbial communities that were associated in maternal-infant dyads at 6 weeks of life and found that milk microbial communities were more strongly associated with infant gut microbial communities in infants delivered operatively and after a lag period. These results suggest that milk microbial communities have a long-term effect on the infant gut microbiome both through sharing of microbes and other molecular mechanisms.
Collapse
Affiliation(s)
- Sara N. Lundgren
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Juliette C. Madan
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
- Department of Pediatrics, Children’s Hospital at Dartmouth, Lebanon, NH, United States
- Department of Psychiatry, Children’s Hospital at Dartmouth, Lebanon, NH, United States
| | - Margaret R. Karagas
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
- Department of Community and Family Medicine, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Hilary G. Morrison
- Josephine Bay Paul Center, Marine Biological Laboratory, Woods Hole, MA, United States
| | - Brock C. Christensen
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
- Department of Community and Family Medicine, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
- *Correspondence: Brock C. Christensen,
| | - Anne G. Hoen
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| |
Collapse
|
44
|
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
|
45
|
Human Milk Microbiome and Microbiome-Related Products: Potential Modulators of Infant Growth. Nutrients 2022; 14:nu14235148. [PMID: 36501178 PMCID: PMC9737635 DOI: 10.3390/nu14235148] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/25/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Infant growth trajectory may influence later-life obesity. Human milk provides a wide range of nutritional and bioactive components that are vital for infant growth. Compared to formula-fed infants, breastfed infants are less likely to develop later-onset obesity, highlighting the potential role of bioactive components present in human milk. Components of particular interest are the human milk microbiota, human milk oligosaccharides (HMOs), short-chain fatty acids (SCFAs), and antimicrobial proteins, each of which influence the infant gut microbiome, which in turn has been associated with infant body composition. SCFAs and antimicrobial proteins from human milk may also systemically influence infant metabolism. Although inconsistent, multiple studies have reported associations between HMOs and infant growth, while studies on other bioactive components in relation to infant growth are sparse. Moreover, these microbiome-related components may interact with each other within the mammary gland. Here, we review the evidence around the impact of human milk microbes, HMOs, SCFAs, and antimicrobial proteins on infant growth. Breastfeeding is a unique window of opportunity to promote optimal infant growth, with aberrant growth trajectories potentially creating short- and long-term public health burdens. Therefore, it is important to understand how bioactive components of human milk influence infant growth.
Collapse
|
46
|
Gonzalez-Visiedo M, Kulis MD, Markusic DM. Manipulating the microbiome to enhance oral tolerance in food allergy. Cell Immunol 2022; 382:104633. [PMID: 36347161 DOI: 10.1016/j.cellimm.2022.104633] [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: 07/15/2022] [Revised: 10/06/2022] [Accepted: 10/18/2022] [Indexed: 01/13/2023]
Abstract
Loss of oral tolerance (OT) to food antigens results in food allergies. One component of achieving OT is the symbiotic microorganisms living in the gut (microbiota). The composition of the microbiota can drive either pro-tolerogenic or pro-inflammatory responses against dietary antigens though interactions with the local immune cells within the gut. Products from bacterial fermentation, such as butyrate, are one of the main communication molecules involved in this interaction, however, this is released by a subset of bacterial species. Thus, strategies to specifically expand these bacteria with protolerogenic properties have been explored to complement oral immunotherapy in food allergy. These approaches either provide digestible biomolecules to induce beneficial bacteria species (prebiotics) or the direct administration of live bacteria species (probiotics). While this combined therapy has shown positive outcomes in clinical trials for cow's milk allergy, more research is needed to determine if this therapy can be extended to other food allergens.
Collapse
Affiliation(s)
- Miguel Gonzalez-Visiedo
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Michael D Kulis
- Department of Pediatrics, Division of Allergy and Immunology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - David M Markusic
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
47
|
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
|
48
|
Overgaard Poulsen K, Astono J, Jakobsen RR, Uldbjerg N, Fuglsang J, Nielsen DS, Sundekilde UK. Influence of maternal body mass index on human milk composition and associations to infant metabolism and gut colonisation: MAINHEALTH - a study protocol for an observational birth cohort. BMJ Open 2022; 12:e059552. [PMID: 36323479 PMCID: PMC9639067 DOI: 10.1136/bmjopen-2021-059552] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
INTRODUCTION Human milk provides all macronutrients for growth, bioactive compounds, micro-organisms and immunological components, which potentially interacts with and primes infant growth and, development, immune responses and the gut microbiota of the new-born. Infants with an overweight mother are more likely to become overweight later in life and overweight has been related to the gut microbiome. Therefore, it is important to investigate the mother-milk-infant triad as a biological system and if the maternal weight status influences the human milk composition, infant metabolism and gut microbiome. METHODS AND ANALYSIS This study aims to include 200 mother-infant dyads stratified into one of three body mass index (BMI) categories based on mother's prepregnancy BMI. Multiomics analyses include metabolomics, proteomics, glycomics and microbiomics methods, aiming to characterise human milk from the mothers and further relate the composition to infant gut microbiota and its metabolic impact in the infant. Infant gut microbiota is analysed using 16S sequencing of faeces samples. Nuclear magnetic resonance and mass spectrometry are used for the remaining omics analysis. We investigate whether maternal pre-pregnancy BMI results in a distinct human milk composition that potentially affects the initial priming of the infant's gut environment and metabolism early in life. ETHICS AND DISSEMINATION The Central Denmark Region Committees on Health Research Ethics has approved the protocol (J-nr. 1-10-72-296-18). All participants have before inclusion signed informed consent and deputy informed consent in accordance with the Declaration of Helsinki II. Results will be disseminated to health professionals including paediatricians, research community, nutritional policymakers, industry and finally the public. The scientific community will be informed via peer-reviewed publications and presentations at scientific conferences, the industry will be invited for meetings, and the public will be informed via reports in science magazines and the general press. Data cleared for personal data, will be deposited at public data repositories. TRIAL REGISTRATION NUMBER Danish regional committee of the Central Jutland Region, journal number: 1-10-72-296-18, version 6.Danish Data Protection Agency, journal number: 2016-051-000001, 1304. CLINICALTRIALS gov, identifier: NCT05111990.
Collapse
Affiliation(s)
- Katrine Overgaard Poulsen
- Department of Food Science, Aarhus University, Aarhus N, Denmark
- Sino-Danish Centre for Education and Research (SDC), University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Julie Astono
- Department of Food Science, Aarhus University, Aarhus N, Denmark
| | | | - Niels Uldbjerg
- Department of Clinical Medicine, Aarhus University Hospital, Aarhus N, Denmark
| | - Jens Fuglsang
- Department of Clinical Medicine, Aarhus University Hospital, Aarhus N, Denmark
| | | | | |
Collapse
|
49
|
Daiy K, Harries V, Nyhan K, Marcinkowska UM. Maternal weight status and the composition of the human milk microbiome: A scoping review. PLoS One 2022; 17:e0274950. [PMID: 36191014 PMCID: PMC9529148 DOI: 10.1371/journal.pone.0274950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 09/07/2022] [Indexed: 11/19/2022] Open
Abstract
The human milk microbiome is thought to partly contribute to the assembly of the infant gut microbiome, a microbial community with important implications for infant health and development. While obesity has well-established links with the adult gut microbiome, less is known about how it affects the human milk microbiome. In this scoping review, we synthesize the current literature on the microbial composition of human milk by maternal weight status, defined broadly as BMI (prepregnancy and postpartum) and gestational weight gain (GWG). This study followed the a priori protocol published in Prospero (registration #: CRD42020165633). We searched the following databases for studies reporting maternal weight status and a characterization of milk microbiota through culture-dependent and culture-independent methods: MEDLINE, Embase, Web of Science, CINAHL, and Scopus. After screening 6,365 studies, we found 20 longitudinal and cross-sectional studies investigating associations between maternal weight status and the composition of the milk microbiome. While some studies reported no associations, many others reported that women with a pre-pregnancy or postpartum BMI characterized as overweight or obese, or with excessive GWG, had higher abundances of the genus Staphylococcus, lower Bifidobacterium abundance, and lower alpha diversity (within-sample diversity). This review suggests that maternal weight status is minorly associated with the composition of the milk microbiome in various ways. We offer potential explanations for these findings, as well as suggestions for future research.
Collapse
Affiliation(s)
- Katherine Daiy
- Department of Anthropology, Yale University, New Haven, CT, United States of America
| | - Victoria Harries
- Department of Anthropology, Yale University, New Haven, CT, United States of America
| | - Kate Nyhan
- Cushing/Whitney Medical Library, Yale University, New Haven, CT, United States of America
| | | |
Collapse
|
50
|
Jordan A, Carding SR, Hall LJ. The early-life gut microbiome and vaccine efficacy. THE LANCET. MICROBE 2022; 3:e787-e794. [PMID: 36088916 DOI: 10.1016/s2666-5247(22)00185-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 05/31/2022] [Accepted: 06/08/2022] [Indexed: 12/24/2022]
Abstract
Vaccines are one of the greatest successes of public health, preventing millions of cases of disease and death in children each year. However, the efficacy of many vaccines can vary greatly between infants from geographically and socioeconomically distinct locations. Differences in the composition of the intestinal microbiome have emerged as one of the main factors that can account for variations in immunisation outcomes. In this Review, we assess the influence of the gut microbiota upon early life immunity, focusing on two important members of the microbiota with health-promoting and immunomodulatory properties: Bifidobacterium and Bacteroides. Additionally, we discuss their immune stimulatory microbial properties, interactions with the host, and their effect on vaccine responses and efficacy in infants. We also provide an overview of current microbiota-based approaches to enhance vaccine outcomes, and describe novel microbe-derived components that could lead to safer, more effective vaccines and vaccine adjuvants.
Collapse
Affiliation(s)
- Anne Jordan
- Gut Microbes & Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Simon R Carding
- Gut Microbes & Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK; Norwich Medical School, University of East Anglia, Norwich, UK
| | - Lindsay J Hall
- Gut Microbes & Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK; Norwich Medical School, University of East Anglia, Norwich, UK; Intestinal Microbiome, School of Life Sciences, ZIEL Institute for Food & Health, Technical University of Munich, Munich, Germany.
| |
Collapse
|