1
|
Costa JSP, Brandão HV, Amaral MVC, Santos GCD, Cruz Martins CD, Ramos MDSX, Vieira TDO, Benevides RG, Vieira GO. Protocol for the collection, packaging, and transportation of preterm newborn stool samples. Clinics (Sao Paulo) 2024; 79:100531. [PMID: 39541673 PMCID: PMC11605465 DOI: 10.1016/j.clinsp.2024.100531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 09/17/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND The newborn's intestinal microbiota can vary with gestational age. Reliable analyses of stool samples require establishing the steps of collection, packaging, transportation, and storage. OBJECTIVE To describe the development of a protocol and test an algorithm for the sequence of actions and procedures for the collection, packaging, transportation, and storage of stool samples from Preterm Newborn (PTNB) admitted to a Neonatal Intensive Care Unit (NICU). MATERIALS AND METHODS This is a descriptive, observational study linked to a non-randomized controlled clinical trial conducted in the NICU of two public hospitals covenanted to the Unified Health System of a large city in inland northeastern Brazil. The methodology adopted in its development followed the Brazilian Ministry of Health Preparation Guide recommendations. RESULTS The flow of actions and procedures was consistent with scientific evidence. The sequence of work process steps to implement the proposed protocol was tested and resulted in a graphical representation of an algorithm compatible with the reality of Brazilian public hospitals. CONCLUSION Following the protocol steps with the description of the rules of conduct and recommendations regarding the collection, packaging, transportation, and storage of stool samples from PTNBs ensured the preservation and integrity of the bacterial DNA in the stools sample. The details of the recommendations will allow their reproducibility and improvement by professionals and researchers with similar study objects. TRIAL REGISTRATION World Health Organization (WHO) under Universal Trial Number (UTN) code U1111-1266-2295, under register RBR-3mm7cs in the Brazilian Registry of Clinical Trials (REBEC).
Collapse
Affiliation(s)
- Jessica Santos Passos Costa
- Health Research and Extension Center, Postgraduate Program in Public Health, Department of Health, Universidade Estadual de Feira de Santana, Feira de Santana, BA, Brazil; PhD in Public Health, Universidade Estadual de Feira de Santana, Feira de Santana, BA, Brazil.
| | - Heli Vieira Brandão
- Health Research and Extension Center, Postgraduate Program in Public Health, Department of Health, Universidade Estadual de Feira de Santana, Feira de Santana, BA, Brazil; PhD in Medicine and Human Health, Escola Baiana de Medicina e Saúde Pública, Salvador, BA, Brazil
| | - Mara Viana Cardoso Amaral
- Health Research and Extension Center, Department of Health, Universidade Federal do Recôncavo da Bahia, Cruz das Almas, BA, Brazil; Master's degree in Public Health, Universidade Estadual de Feira de Santana, Feira de Santana, BA, Brazil
| | - Gabriela Cintra Dos Santos
- Health Research and Extension Center, Postgraduate Program in Public Health, Department of Health, Universidade Estadual de Feira de Santana, Feira de Santana, BA, Brazil; Master's degree in Public Health, Universidade Estadual de Feira de Santana, Feira de Santana, BA, Brazil
| | - Camilla da Cruz Martins
- Health Research and Extension Center, Postgraduate Program in Public Health, Department of Health, Universidade Estadual de Feira de Santana, Feira de Santana, BA, Brazil; PhD in Public Health, Universidade Estadual de Feira de Santana, Feira de Santana, BA, Brazil
| | - Michelle de Santana Xavier Ramos
- Health Research and Extension Center, Postgraduate Program in Public Health, Department of Health, Universidade Estadual de Feira de Santana, Feira de Santana, BA, Brazil; PhD in Public Health, Universidade Estadual de Feira de Santana, Feira de Santana, BA, Brazil
| | - Tatiana de Oliveira Vieira
- Health Research and Extension Center, Postgraduate Program in Public Health, Department of Health, Universidade Estadual de Feira de Santana, Feira de Santana, BA, Brazil; PhD in Medicine and Health, Universidade Federal da Bahia, Salvador, BA, Brazil
| | - Raquel Guimarães Benevides
- Department of Biology, Universidade Estadual de Feira de Santana, Feira de Santana, BA, Brazil; PhD in Biologie Structurale et Nanobiologie, Université Joseph Fourier ‒ Grenoble I, France
| | - Graciete Oliveira Vieira
- Health Research and Extension Center, Postgraduate Program in Public Health, Department of Health, Universidade Estadual de Feira de Santana, Feira de Santana, BA, Brazil; PhD in Medicine and Health, Universidade Federal da Bahia, Salvador, BA, Brazil
| |
Collapse
|
2
|
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
|
3
|
Masi AC, Beck LC, Perry JD, Granger CL, Hiorns A, Young GR, Bode L, Embleton ND, Berrington JE, Stewart CJ. Human milk microbiota, oligosaccharide profiles, and infant gut microbiome in preterm infants diagnosed with necrotizing enterocolitis. Cell Rep Med 2024; 5:101708. [PMID: 39216480 PMCID: PMC11524953 DOI: 10.1016/j.xcrm.2024.101708] [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: 04/10/2024] [Revised: 06/25/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
Necrotizing enterocolitis (NEC) is a severe intestinal disease of very preterm infants with mother's own milk (MOM) providing protection, but the contribution of the MOM microbiota to NEC risk has not been explored. Here, we analyze MOM of 110 preterm infants (48 NEC, 62 control) in a cross-sectional study. Breast milk contains viable bacteria, but there is no significant difference in MOM microbiota between NEC and controls. Integrative analysis between MOM microbiota, human milk oligosaccharides (HMOs), and the infant gut microbiota shows positive correlations only between Acinetobacter in the infant gut and Acinetobacter and Staphylococcus in MOM. This study suggests that NEC protection from MOM is not modulated through the MOM microbiota. Thus, "'restoring" the MOM microbiota in donor human milk is unlikely to reduce NEC, and emphasis should instead focus on increasing fresh maternal human milk intake and researching different therapies for NEC prevention.
Collapse
Affiliation(s)
- Andrea C Masi
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Lauren C Beck
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - John D Perry
- Microbiology Department, Freeman Hospital, Newcastle upon Tyne NE7 7DN, UK
| | - Claire L Granger
- Newcastle Neonatal Service, Newcastle Hospitals NHS Trust, Newcastle upon Tyne NE1 4LP, UK
| | - Alice Hiorns
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Gregory R Young
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Lars Bode
- Department of Pediatrics, Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (MOMI CORE), University of California San Diego, La Jolla, CA 92093, USA; The Human Milk Institute (HMI), University of California San Diego, La Jolla, CA 92093, USA
| | - Nicholas D Embleton
- Newcastle Neonatal Service, Newcastle Hospitals NHS Trust, Newcastle upon Tyne NE1 4LP, UK; Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Janet E Berrington
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; Newcastle Neonatal Service, Newcastle Hospitals NHS Trust, Newcastle upon Tyne NE1 4LP, UK.
| | - Christopher J Stewart
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK.
| |
Collapse
|
4
|
Shama S, Asbury MR, O'Connor DL. From parent to progeny. Cell Host Microbe 2024; 32:947-949. [PMID: 38870905 DOI: 10.1016/j.chom.2024.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 06/15/2024]
Abstract
How infants acquire their gut microbial communities and the various factors influencing these dynamics remain unclear. In this issue of Cell Host & Microbe, Selma-Royo et al. and Dubois et al. use shotgun metagenomic sequencing to understand the transmission of microbes from parents to infants and delve into factors modifying this process.
Collapse
Affiliation(s)
- Sara Shama
- Department of Nutritional Sciences, University of Toronto, Toronto, Canada; Translational Medicine Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, Canada
| | - Michelle R Asbury
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Canada; Department of Pediatrics, University of Calgary, Calgary, Canada; Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Canada; Alberta Children's Hospital Research Institute, Calgary, Canada
| | - Deborah L O'Connor
- Department of Nutritional Sciences, University of Toronto, Toronto, Canada; Translational Medicine Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, Canada; Joannah and Brian Lawson Centre for Child Nutrition, University of Toronto, Toronto, Canada; Rogers Hixon Ontario Human Milk Bank, Toronto, Canada.
| |
Collapse
|
5
|
Younge N. Influence of infant microbiome on health and development. Clin Exp Pediatr 2024; 67:224-231. [PMID: 37605538 PMCID: PMC11065641 DOI: 10.3345/cep.2023.00598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/08/2023] [Accepted: 07/19/2023] [Indexed: 08/23/2023] Open
Abstract
The microbiome is a complex ecosystem comprising microbes, their genomes, and the surrounding environment. The microbiome plays a critical role in early human development, including maturation of the host immune system and gastrointestinal tract. Multiple factors, including diet, anti-biotic use, and other environmental exposures, influence the establishment of the microbiome during infancy. Numerous studies have identified associations between the microbiome and neonatal diseases, including necrotizing enterocolitis, sepsis, and malnutrition. Furthermore, there is compelling evidence that perturbation of the microbiome in early life can have lasting developmental effects that increase an individual's risk for immune and metabolic diseases in later life. Supplementation of the microbiome with probiotics reduces the risk of necrotizing enterocolitis and sepsis in at-risk infants. This review focuses on the structure and function of the infant microbiome, the environmental and clinical factors that influence its assembly, and its impact on infant health and development.
Collapse
|
6
|
Gao Y, Lu X, Pan M, Liu C, Min Y, Chen X. Effect of breast milk intake volume on early behavioral neurodevelopment of extremely preterm infants. Int Breastfeed J 2024; 19:3. [PMID: 38233943 PMCID: PMC10795355 DOI: 10.1186/s13006-024-00612-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 01/01/2024] [Indexed: 01/19/2024] Open
Abstract
BACKGROUND This study aimed to explore the effects of breast milk feeding volume on the early behavioral neurodevelopment of extremely preterm infants (gestational age < 28 weeks). METHODS The study was conducted from 1 January 2021 to 31 March 2023. A total of 187 preterm infants from a neonatal intensive care unit (NICU) in a Grade III Class A hospital in Zhejiang, China, were divided based on the proportion of breast milk in their total enteral nutrition: high proportion (≥ 80%, including exclusive breast milk feeding), medium proportion (20% ~ < 80%), and low proportion (< 20%). The study investigated motor performance and behavioral neurodevelopment at 37 weeks of corrected gestational age, as well as the total incidence of intracranial hemorrhage within the first four weeks postpartum. RESULTS The low breast milk feeding group had significantly lower scores in infant motor performance (31.34 ± 5.85) and elicited item scores (19.89 ± 5.55) compared to the medium and high groups (33.52 ± 4.33, 22.13 ± 4.22; and 35.86 ± 5.27, 23.91 ± 4.98), p < 0.05, respectively. Despite no significant difference in behavioral ability, the low proportion group exhibited lower passive muscle tension and primitive reflex scores than the medium and high proportion groups. The high proportion group showed higher active muscle tension scores. Ultrasound results revealed varying incidences of intracranial hemorrhage: 72.9% in low, 52.5% in medium, and 19.6% in the high proportion groups. CONCLUSIONS Medium to high levels of breast milk feeding contribute positively to motor and behavioral neurological development in extremely preterm infants and decrease the likelihood of ventricular hemorrhage. However, it does not have a significant effect on the development of behavioral abilities. Due to the limited sample size, the next step will be to expand the sample size and further investigate the extent of the impact on various aspects of the nervous system.
Collapse
Affiliation(s)
- Ying Gao
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaoyu Lu
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Mengqing Pan
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chuntian Liu
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yuxiao Min
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaochun Chen
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
7
|
Filatava EJ, Liu Z, Xie J, Tran DB, Chen K, El Habbal N, Weinstock G, Zhou Y, Gregory KE. The preterm human milk microbiota fluctuates by postpartum week and is characterized by gestational age and maternal BMI. mBio 2023; 14:e0210623. [PMID: 37975676 PMCID: PMC10746270 DOI: 10.1128/mbio.02106-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 09/28/2023] [Indexed: 11/19/2023] Open
Abstract
IMPORTANCE Despite a growing recognition that the type of nutrition received by preterm infants influences their intestinal microbiome and health outcomes, the microbiota of mother's own milk (MOM), pasteurized donor human milk (PDHM), and infant formula remain poorly characterized. In our study, we found that the structure of microbial communities, bacterial diversity, and relative abundances of specific genera were significantly different between MOM, PDHM, and formula. Additionally, our results suggest that the microbiota of MOM changes as a function of time and maternal factors. Lastly, we identified three lactotypes within MOM that have distinct microbial compositions and described the maternal factors associated with them. These findings set the stage for future research aimed at advancing our knowledge of the microbiota of preterm infant nutrition and the specific influence it may have on health outcomes.
Collapse
Affiliation(s)
| | - Zhongmao Liu
- University of Connecticut, Storrs, Connecticut, USA
| | - Jiaojiao Xie
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | | | - Kun Chen
- University of Connecticut, Storrs, Connecticut, USA
| | | | | | - Yanjiao Zhou
- University of Connecticut, Storrs, Connecticut, USA
| | | |
Collapse
|
8
|
Singh P, Al Mohannadi N, Murugesan S, Almarzooqi F, Kabeer BSA, Marr AK, Kino T, Brummaier T, Terranegra A, McGready R, Nosten F, Chaussabel D, Al Khodor S. Unveiling the dynamics of the breast milk microbiome: impact of lactation stage and gestational age. J Transl Med 2023; 21:784. [PMID: 37932773 PMCID: PMC10629158 DOI: 10.1186/s12967-023-04656-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 10/24/2023] [Indexed: 11/08/2023] Open
Abstract
BACKGROUND Breast milk (BM) provides complete nutrition for infants for the first six months of life and is essential for the development of the newborn's immature immune and digestive systems. While BM was conventionally believed to be sterile, recent advanced high throughput technologies have unveiled the presence of diverse microbial communities in BM. These insights into the BM microbiota have mainly originated from uncomplicated pregnancies, possibly not reflecting the circumstances of mothers with pregnancy complications like preterm birth (PTB). METHODS In this article, we investigated the BM microbial communities in mothers with preterm deliveries (before 37 weeks of gestation). We compared these samples with BM samples from healthy term pregnancies across different lactation stages (colostrum, transitional and mature milk) using 16S rRNA gene sequencing. RESULTS Our analysis revealed that the microbial communities became increasingly diverse and compositionally distinct as the BM matured. Specifically, mature BM samples were significantly enriched in Veillonella and lactobacillus (Kruskal Wallis; p < 0.001) compared to colostrum. The comparison of term and preterm BM samples showed that the community structure was significantly different between the two groups (Bray Curtis and unweighted unifrac dissimilarity; p < 0.001). Preterm BM samples exhibited increased species richness with significantly higher abundance of Staphylococcus haemolyticus, Propionibacterium acnes, unclassified Corynebacterium species. Whereas term samples were enriched in Staphylococcus epidermidis, unclassified OD1, and unclassified Veillonella among others. CONCLUSION Our study underscores the significant influence of pregnancy-related complications, such as preterm birth (before 37 weeks of gestation), on the composition and diversity of BM microbiota. Given the established significance of the maternal microbiome in shaping child health outcomes, this investigation paves the way for identifying modifiable factors that could optimize the composition of BM microbiota, thereby promoting maternal and infant health.
Collapse
Affiliation(s)
- Parul Singh
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
- Research Department, Sidra Medicine, Doha, Qatar
| | | | | | | | | | | | | | - Tobias Brummaier
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | | | - Rose McGready
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - François Nosten
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Damien Chaussabel
- Research Department, Sidra Medicine, Doha, Qatar
- The Jackson Laboratories, Farmington, CT, USA
| | - Souhaila Al Khodor
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar.
- Research Department, Sidra Medicine, Doha, Qatar.
| |
Collapse
|
9
|
Xiang Q, Yan X, Shi W, Li H, Zhou K. Early gut microbiota intervention in premature infants: Application perspectives. J Adv Res 2023; 51:59-72. [PMID: 36372205 PMCID: PMC10491976 DOI: 10.1016/j.jare.2022.11.004] [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: 08/08/2022] [Revised: 10/30/2022] [Accepted: 11/05/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Preterm birth is the leading cause of death in children under the age of five. One of the major factors contributing to the high risk of diseases and deaths in premature infants is the incomplete development of the intestinal immune system. The gut microbiota has been widely recognized as a critical factor in promoting the development and function of the intestinal immune system after birth. However, the gut microbiota of premature infants is at high risk of dysbiosis, which is highly associated with adverse effects on the development and education of the early life immune system. Early intervention can modulate the colonization and development of gut microbiota and has a long-term influence on the development of the intestinal immune system. AIM OF REVIEW This review aims to summarize the characterization, interconnection, and underlying mechanism of gut microbiota and intestinal innate immunity in premature infants, and to discuss the status, applicability, safety, and prospects of different intervention strategies in premature infants, thus providing an overview and outlook of the current applications and remaining gaps of early intervention strategies in premature infants. KEY SCIENTIFIC CONCEPTS OF REVIEW This review is focused on three key concepts. Firstly, the gut microbiota of premature infants is at high risk of dysbiosis, resulting in dysfunctional intestinal immune system processes. Secondly, contributing roles of early intervention have been observed in improving the intestinal environment and promoting gut microbiota colonization, which is significant in the development and function of gut immunity in premature infants. Thirdly, different strategies of early intervention, such as probiotics, fecal microbiota transplantation, and nutrients, show different safety, applicability, and outcome in premature infants, and the underlying mechanism is complex and poorly understood.
Collapse
Affiliation(s)
- Quanhang Xiang
- Shenzhen Institute of Respiratory Diseases, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Xudong Yan
- Department of Neonatal Intensive Care Unit, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Wei Shi
- Department of Obstetrics and Gynecology, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Huiping Li
- Department of Respiratory and Critical Care Medicine, the first affiliated hospital of Southern University of Science and Technology of China, Shenzhen People's Hospital, Shenzhen, China; The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China
| | - Kai Zhou
- Shenzhen Institute of Respiratory Diseases, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China; The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
10
|
Miura K, Tanaka M, Date M, Ito M, Mizuno N, Mizuno K. Comparison of bacterial profiles in human milk from mothers of term and preterm infants. Int Breastfeed J 2023; 18:29. [PMID: 37291566 DOI: 10.1186/s13006-023-00563-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 05/14/2023] [Indexed: 06/10/2023] Open
Abstract
BACKGROUND Reducing the disposal of donated human milk (HM) is important for efficient management of human milk banks (HMBs). The presence of bacteria growth is the main factor that contributes to the disposal of donated HM. The bacterial profile in HM is suspected to differ between term and preterm mothers, with HM from preterm mothers containing more bacteria. Thus, elucidation of the causes of bacterial growth in preterm and term HM may help to reduce the disposal of donated preterm HM. This study compared the bacterial profiles of HM between mothers of term infants and mothers of preterm infants. METHODS This pilot study was conducted in the first Japanese HMB, which was initiated in 2017. This study analyzed 214 human milk samples (term: 75, preterm: 139) donated by 47 registered donors (term: 31, preterm: 16) from January to November 2021. Bacterial culture results in term and preterm HM were retrospectively reviewed in May 2022. Differences in total bacterial count and bacterial species count per batch were analyzed using the Mann-Whitney U test. Bacterial loads were analyzed using the Chi-square test or Fisher's exact test. RESULTS The disposal rate did not significantly differ between term and preterm groups (p = 0.77), but the total amount of disposal was greater in the preterm group (p < 0.01). Coagulase-negative Staphylococci, Staphylococcus aureus, and Pseudomonas fluorescens were frequently found in both types of HM. Serratia liquefaciens (p < 0.001) and two other bacteria were present in term HM; a total of five types of bacteria, including Enterococcus faecalis and Enterobacter aerogenes (p < 0.001) were present in preterm HM. The median (interquartile range) total bacterial counts were 3,930 (435-23,365) colony-forming units (CFU)/mL for term HM and 26,700 (4,050-334,650) CFU/mL for preterm HM (p < 0.001). CONCLUSIONS This study revealed that HM from preterm mothers had a higher total bacterial count and different types of bacteria than HM from term mothers. Additionally, preterm infants can receive nosocomial-infection-causing bacteria in the NICU through their mother's milk. Enhanced hygiene instructions for preterm mothers may reduce the disposal of valuable preterm human milk, along with the risk of HM pathogen transmission to infants in NICUs.
Collapse
Affiliation(s)
- Kumiko Miura
- The Nippon Foundation Human Milk Bank, Nihonbashi-koamicho Square Building 1F, 17-10 Nihonbashi-koamicho, Chuo-ku, Tokyo, 103-0016, Japan
- Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-machi, Yufu-shi, Oita, 879-5593, Japan
| | - Miori Tanaka
- The Nippon Foundation Human Milk Bank, Nihonbashi-koamicho Square Building 1F, 17-10 Nihonbashi-koamicho, Chuo-ku, Tokyo, 103-0016, Japan
| | - Midori Date
- The Nippon Foundation Human Milk Bank, Nihonbashi-koamicho Square Building 1F, 17-10 Nihonbashi-koamicho, Chuo-ku, Tokyo, 103-0016, Japan
| | - Mizuho Ito
- The Nippon Foundation Human Milk Bank, Nihonbashi-koamicho Square Building 1F, 17-10 Nihonbashi-koamicho, Chuo-ku, Tokyo, 103-0016, Japan
| | - Noriko Mizuno
- Japan Human Milk Bank Association, 4-4 Nihonbashi-Hisamatsucho, Chuo-ku, Tokyo, 103-8480, Japan
| | - Katsumi Mizuno
- The Nippon Foundation Human Milk Bank, Nihonbashi-koamicho Square Building 1F, 17-10 Nihonbashi-koamicho, Chuo-ku, Tokyo, 103-0016, Japan.
- Japan Human Milk Bank Association, 4-4 Nihonbashi-Hisamatsucho, Chuo-ku, Tokyo, 103-8480, Japan.
- Department of Pediatrics, Showa University of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8666, Japan.
| |
Collapse
|
11
|
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
|
12
|
Abdolmaleky HM, Martin M, Zhou JR, Thiagalingam S. Epigenetic Alterations of Brain Non-Neuronal Cells in Major Mental Diseases. Genes (Basel) 2023; 14:896. [PMID: 37107654 PMCID: PMC10137903 DOI: 10.3390/genes14040896] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/22/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
The tissue-specific expression and epigenetic dysregulation of many genes in cells derived from the postmortem brains of patients have been reported to provide a fundamental biological framework for major mental diseases such as autism, schizophrenia, bipolar disorder, and major depression. However, until recently, the impact of non-neuronal brain cells, which arises due to cell-type-specific alterations, has not been adequately scrutinized; this is because of the absence of techniques that directly evaluate their functionality. With the emergence of single-cell technologies, such as RNA sequencing (RNA-seq) and other novel techniques, various studies have now started to uncover the cell-type-specific expression and DNA methylation regulation of many genes (e.g., TREM2, MECP2, SLC1A2, TGFB2, NTRK2, S100B, KCNJ10, and HMGB1, and several complement genes such as C1q, C3, C3R, and C4) in the non-neuronal brain cells involved in the pathogenesis of mental diseases. Additionally, several lines of experimental evidence indicate that inflammation and inflammation-induced oxidative stress, as well as many insidious/latent infectious elements including the gut microbiome, alter the expression status and the epigenetic landscapes of brain non-neuronal cells. Here, we present supporting evidence highlighting the importance of the contribution of the brain's non-neuronal cells (in particular, microglia and different types of astrocytes) in the pathogenesis of mental diseases. Furthermore, we also address the potential impacts of the gut microbiome in the dysfunction of enteric and brain glia, as well as astrocytes, which, in turn, may affect neuronal functions in mental disorders. Finally, we present evidence that supports that microbiota transplantations from the affected individuals or mice provoke the corresponding disease-like behavior in the recipient mice, while specific bacterial species may have beneficial effects.
Collapse
Affiliation(s)
- Hamid Mostafavi Abdolmaleky
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA;
- Department of Surgery, Nutrition/Metabolism Laboratory, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Marian Martin
- Department of Neurology, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Jin-Rong Zhou
- Department of Surgery, Nutrition/Metabolism Laboratory, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Sam Thiagalingam
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA;
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
13
|
Krebs NF, Belfort MB, Meier PP, Mennella JA, O'Connor DL, Taylor SN, Raiten DJ. Infant factors that impact the ecology of human milk secretion and composition-a report from "Breastmilk Ecology: Genesis of Infant Nutrition (BEGIN)" Working Group 3. Am J Clin Nutr 2023; 117 Suppl 1:S43-S60. [PMID: 37173060 PMCID: PMC10356564 DOI: 10.1016/j.ajcnut.2023.01.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 12/08/2022] [Accepted: 01/03/2023] [Indexed: 05/15/2023] Open
Abstract
Infants drive many lactation processes and contribute to the changing composition of human milk through multiple mechanisms. This review addresses the major topics of milk removal; chemosensory ecology for the parent-infant dyad; the infant's inputs into the composition of the human milk microbiome; and the impact of disruptions in gestation on the ecology of fetal and infant phenotypes, milk composition, and lactation. Milk removal, which is essential for adequate infant intake and continued milk synthesis through multiple hormonal and autocrine/paracrine mechanisms, should be effective, efficient, and comfortable for both the lactating parent and the infant. All 3 components should be included in the evaluation of milk removal. Breastmilk "bridges" flavor experiences in utero with postweaning foods, and the flavors become familiar and preferred. Infants can detect flavor changes in human milk resulting from parental lifestyle choices, including recreational drug use, and early experiences with the sensory properties of these recreational drugs impact subsequent behavioral responses. Interactions between the infant's own developing microbiome, that of the milk, and the multiple environmental factors that are drivers-both modifiable and nonmodifiable-in the microbial ecology of human milk are explored. Disruptions in gestation, especially preterm birth and fetal growth restriction or excess, impact the milk composition and lactation processes such as the timing of secretory activation, adequacy of milk volume and milk removal, and duration of lactation. Research gaps are identified in each of these areas. To assure a sustained and robust breastfeeding ecology, these myriad infant inputs must be systematically considered.
Collapse
Affiliation(s)
- Nancy F Krebs
- Section of Nutrition, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA.
| | - Mandy B Belfort
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Paula P Meier
- Department of Pediatrics, Rush University Medical Center, Chicago, IL, USA
| | | | - Deborah L O'Connor
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada; The Hospital for Sick Children, Toronto, ON, Canada
| | - Sarah N Taylor
- Division of Neonatology, Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | - Daniel J Raiten
- Pediatric Growth and Nutrition Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
14
|
Differences in the Microbiological Profile of Raw and Pasteurized Breastmilk from Hospital and Community-Based Donors at the First Human Milk Bank in Vietnam. Nutrients 2023; 15:nu15020412. [PMID: 36678283 PMCID: PMC9864883 DOI: 10.3390/nu15020412] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/06/2023] [Accepted: 01/11/2023] [Indexed: 01/15/2023] Open
Abstract
Background: Microbiological quality is one of the key safety standards in human milk bank (HMB) operations. We describe the profiles of bacteria in donor human milk (DHM) before and after the pasteurization of samples collected from breastfeeding women in the hospital and from the community in the first HMB in Vietnam. Methods: Data were collected between February 2017 and January 2022 from an online HMB monitoring system. First, DHM samples were cultured, and the number of colony-forming units (CFU) were counted before (n = 708) and after pasteurization (n = 1146). The gram-staining method combined with the Vitek 2 Compact system were used to identify types of organisms at the Da Nang Hospital for Women and Children’s Laboratory. Passing criteria for DHM included pre-pasteurization samples had a total colony count <105 CFU/mL and post-pasteurization was <10 CFU/mL. Results: During five years of operation, Da Nang HMB had 491 donors (48.7% were hospital and the rest community donors) who donated an average amount of 14.2 L over 45 days. Of this DHM volume, 84.9% of donor samples passed the pre- and post-pasteurization microbiological tests. DHM from community donors had a higher pass rate (87.8%) compared to that from hospital donors (79.5%). Before pasteurization, 15.4% of DHM samples had a bacteria count <103 CFU/mL, 63.0% had 103-<105 CFU/mL, and 21.6% had ≥105 CFU/mL. Most of the unpasteurized DHM samples (93.0%) had microorganism growth: with one organism (16.4%), two (33.9%), three or more (43.6%). After pasteurization, 17.9% samples had a bacteria count of 1−9 CFU/mL and 7.2% had ≥10 CFU/mL. DHM samples from community donors had a lower bacterial count and number of organisms than those from hospital donors both before and after pasteurization. The highest microorganisms from unpasteurized DHM samples were Staphylococcus epidermidis (74.2%), Acinetobacter sp. (52.1%), gram-positive bacillus (51.7%), Staphylococcus coagulase-negative (15.8%), and Staphylococcus aureus (10.5%). Common microorganisms from pasteurized DHM were gram-positive bacillus (21.0%), Staphylococcus epidermidis (3.9%), and Acinetobacter sp. (0.9%). Samples from the hospital tended to have a higher contamination with those microorganisms than those from community donors. Conclusions: The majority of DHM samples in Da Nang passed microbiological testing criteria. DHM from community donors had higher pass rates than hospital donors. Corrective actions are needed to improve HMB operations and hospital microbiological quality standards, as well as general improvements in water and sanitation.
Collapse
|
15
|
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
|
16
|
Maternal weight status and the composition of the human milk microbiome: A scoping review. PLoS One 2022; 17:e0274950. [PMID: 36191014 PMCID: PMC9529148 DOI: 10.1371/journal.pone.0274950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 09/07/2022] [Indexed: 11/19/2022] Open
Abstract
The human milk microbiome is thought to partly contribute to the assembly of the infant gut microbiome, a microbial community with important implications for infant health and development. While obesity has well-established links with the adult gut microbiome, less is known about how it affects the human milk microbiome. In this scoping review, we synthesize the current literature on the microbial composition of human milk by maternal weight status, defined broadly as BMI (prepregnancy and postpartum) and gestational weight gain (GWG). This study followed the a priori protocol published in Prospero (registration #: CRD42020165633). We searched the following databases for studies reporting maternal weight status and a characterization of milk microbiota through culture-dependent and culture-independent methods: MEDLINE, Embase, Web of Science, CINAHL, and Scopus. After screening 6,365 studies, we found 20 longitudinal and cross-sectional studies investigating associations between maternal weight status and the composition of the milk microbiome. While some studies reported no associations, many others reported that women with a pre-pregnancy or postpartum BMI characterized as overweight or obese, or with excessive GWG, had higher abundances of the genus Staphylococcus, lower Bifidobacterium abundance, and lower alpha diversity (within-sample diversity). This review suggests that maternal weight status is minorly associated with the composition of the milk microbiome in various ways. We offer potential explanations for these findings, as well as suggestions for future research.
Collapse
|
17
|
Xiao R, Luo G, Liao W, Chen S, Han S, Liang S, Lin Y. Association of human gut microbiota composition and metabolic functions with Ficus hirta Vahl dietary supplementation. NPJ Sci Food 2022; 6:45. [PMID: 36167833 PMCID: PMC9515076 DOI: 10.1038/s41538-022-00161-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 09/09/2022] [Indexed: 12/16/2022] Open
Abstract
Ficus hirta Vahl (FHV), a traditional herbal ingredient of the tonic diet, receives increasing popularity in southern China. However, it is largely unknown that how a FHV diet (FHVD) affects the human gut microbiome. In this exploratory study, a total of 43 healthy individuals were randomized into the FHVD (n = 25) and Control (n = 18) groups to receive diet intervention for 8 weeks. 16S rRNA gene sequencing, metagenomic sequencing and metabolic profile of participants were measured to assess the association between FHV diet and gut microbiome. A preservation effect of Faecalibacterium and enrichment of Dialister, Veillonella, Clostridium, and Lachnospiraceae were found during the FHVD. Accordingly, the pathway of amino acid synthesis, citrate cycle, coenzyme synthesis, and partial B vitamin synthesis were found to be more abundant in the FHVD. In addition, serine, glutamine, gamma-aminobutyric acid, tryptamine, and short-chain fatty acids (SCFAs) were higher after the FHVD. The conjoint analysis of FHV components and in-vitro fermentation confirmed that the improved SCFAs concentration was collectively contributed by the increasing abundance of key enzyme genes and available substrates. In conclusion, the muti-omics analysis showed that the FHVD optimized the structure of the gut microbial community and its metabolic profile, leading to a healthy tendency, with a small cluster of bacteria driving the variation rather than a single taxon.
Collapse
Affiliation(s)
- Ruiming Xiao
- South China University of Technology South China Univ Technol, School of Biology & Biological Engineering, Guangzhou, China.,Guangdong Key Lab Fermentation & Enzyme Engineering, Guangzhou, 510006, China
| | - Guangjuan Luo
- South China University of Technology South China Univ Technol, School of Biology & Biological Engineering, Guangzhou, China.,Guangdong Key Lab Fermentation & Enzyme Engineering, Guangzhou, 510006, China
| | - Wanci Liao
- South China University of Technology South China Univ Technol, School of Biology & Biological Engineering, Guangzhou, China.,Guangdong Key Lab Fermentation & Enzyme Engineering, Guangzhou, 510006, China
| | - Shuting Chen
- South China University of Technology South China Univ Technol, School of Biology & Biological Engineering, Guangzhou, China.,Guangdong Key Lab Fermentation & Enzyme Engineering, Guangzhou, 510006, China
| | - Shuangyan Han
- South China University of Technology South China Univ Technol, School of Biology & Biological Engineering, Guangzhou, China.,Guangdong Key Lab Fermentation & Enzyme Engineering, Guangzhou, 510006, China
| | - Shuli Liang
- South China University of Technology South China Univ Technol, School of Biology & Biological Engineering, Guangzhou, China.,Guangdong Key Lab Fermentation & Enzyme Engineering, Guangzhou, 510006, China
| | - Ying Lin
- South China University of Technology South China Univ Technol, School of Biology & Biological Engineering, Guangzhou, China. .,Guangdong Key Lab Fermentation & Enzyme Engineering, Guangzhou, 510006, China.
| |
Collapse
|
18
|
Kumbhare SV, Jones WD, Fast S, Bonner C, Jong G', Van Domselaar G, Graham M, Narvey M, Azad MB. Source of human milk (mother or donor) is more important than fortifier type (human or bovine) in shaping the preterm infant microbiome. Cell Rep Med 2022; 3:100712. [PMID: 36029771 PMCID: PMC9512671 DOI: 10.1016/j.xcrm.2022.100712] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/13/2022] [Accepted: 07/14/2022] [Indexed: 12/20/2022]
Abstract
Milk fortifiers help meet the nutritional needs of preterm infants receiving their mother's own milk (MOM) or donor human milk. We conducted a randomized clinical trial (NCT03214822) in 30 very low birth weight premature neonates comparing bovine-derived human milk fortifier (BHMF) versus human-derived fortifier (H2MF). We found that fortifier type does not affect the overall microbiome, although H2MF infants were less often colonized by an unclassified member of Clostridiales Family XI. Secondary analyses show that MOM intake is strongly associated with weight gain and microbiota composition, including Bifidobacterium, Veillonella, and Propionibacterium enrichment. Finally, we show that while oxidative stress (urinary F2-isoprostanes) is not affected by fortifier type or MOM intake, fecal calprotectin is higher in H2MF infants and lower in those consuming more MOM. Overall, the source of human milk (mother versus donor) appears more important than the type of milk fortifier (human versus bovine) in shaping preterm infant gut microbiota.
Collapse
Affiliation(s)
- Shreyas V Kumbhare
- Manitoba Interdisciplinary Lactation Centre (MILC), Winnipeg, MB, Canada; Children's Hospital Research Institute of Manitoba (CHRIM), Winnipeg, MB, Canada; Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
| | - William-Diehl Jones
- Children's Hospital Research Institute of Manitoba (CHRIM), Winnipeg, MB, Canada; Faculty of Health Disciplines, Athabasca University, Athabasca, AB, Canada
| | - Sharla Fast
- Department of Nutrition and Food Services, Health Sciences Centre, Winnipeg, MB, Canada
| | - Christine Bonner
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Geert 't Jong
- Children's Hospital Research Institute of Manitoba (CHRIM), Winnipeg, MB, Canada; Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
| | - Gary Van Domselaar
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada; Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - Morag Graham
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada; Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - Michael Narvey
- Children's Hospital Research Institute of Manitoba (CHRIM), Winnipeg, MB, Canada; Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
| | - Meghan B Azad
- Manitoba Interdisciplinary Lactation Centre (MILC), Winnipeg, MB, Canada; Children's Hospital Research Institute of Manitoba (CHRIM), Winnipeg, MB, Canada; Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
19
|
Asbury MR, Shama S, Sa JY, Bando N, Butcher J, Comelli EM, Copeland JK, Forte V, Kiss A, Sherman PM, Stintzi A, Taibi A, Tomlinson C, Unger S, Wang PW, O'Connor DL. Human milk nutrient fortifiers alter the developing gastrointestinal microbiota of very-low-birth-weight infants. Cell Host Microbe 2022; 30:1328-1339.e5. [PMID: 35987195 DOI: 10.1016/j.chom.2022.07.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/14/2022] [Accepted: 07/15/2022] [Indexed: 11/30/2022]
Abstract
Nutrient fortifiers are added to human milk to support the development of very-low-birth-weight infants. Currently, bovine-milk-based fortifiers (BMBFs) are predominantly administered, with increasing interest in adopting human-milk-based fortifiers (HMBFs). Although beneficial for growth, their effects on the gastrointestinal microbiota are unclear. This triple-blind, randomized clinical trial (NCT02137473) tested how nutrient-enriching human milk with HMBF versus BMBF affects the gastrointestinal microbiota of infants born < 1,250 g during hospitalization. HMBF-fed infants (n = 63, n = 269 stools) showed lower microbial diversity, altered microbial community structure, and changes in predicted microbial functions compared with BMBF-fed infants (n = 56, n = 239 stools). HMBF-fed infants had higher relative and normalized abundances of unclassified Enterobacteriaceae and lower abundances of Clostridium sensu stricto. Post hoc analyses identified dose-dependent relationships between individual feed components (volumes of mother's milk, donor milk, and fortifiers) and the microbiota. These results highlight how nutrient fortifiers impact the microbiota of very-low-birth-weight infants during a critical developmental window.
Collapse
Affiliation(s)
- Michelle R Asbury
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Translational Medicine Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Sara Shama
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Translational Medicine Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Jong Yup Sa
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Translational Medicine Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Nicole Bando
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Translational Medicine Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - James Butcher
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1N 6N5, Canada; Ottawa Institute of Systems Biology, University of Ottawa, Ottawa ON, K1H 8M5, Canada
| | - Elena M Comelli
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Joannah and Brian Lawson Centre for Child Nutrition, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Julia K Copeland
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, ON M5S 3B2, Canada
| | - Victoria Forte
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Translational Medicine Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Alex Kiss
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON M5T 3M6, Canada; Evaluative and Clinical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - Philip M Sherman
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Cell Biology Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Gastroenterology, Hepatology, and Nutrition, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1X3, Canada
| | - Alain Stintzi
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1N 6N5, Canada; Ottawa Institute of Systems Biology, University of Ottawa, Ottawa ON, K1H 8M5, Canada
| | - Amel Taibi
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Christopher Tomlinson
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Translational Medicine Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Joannah and Brian Lawson Centre for Child Nutrition, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Pediatrics, University of Toronto, Toronto, ON M5G 1X8, Canada; Division of Neonatology, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Sharon Unger
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Pediatrics, University of Toronto, Toronto, ON M5G 1X8, Canada; Division of Neonatology, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; Department of Pediatrics, Sinai Health, Toronto, ON M5G 1X5, Canada; Rogers Hixon Ontario Human Milk Bank, Toronto, ON M5G 1X5, Canada
| | - Pauline W Wang
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, ON M5S 3B2, Canada
| | - Deborah L O'Connor
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Translational Medicine Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Joannah and Brian Lawson Centre for Child Nutrition, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Pediatrics, Sinai Health, Toronto, ON M5G 1X5, Canada; Rogers Hixon Ontario Human Milk Bank, Toronto, ON M5G 1X5, Canada.
| | | |
Collapse
|
20
|
Orchanian SB, Gauglitz JM, Wandro S, Weldon KC, Doty M, Stillwell K, Hansen S, Jiang L, Vargas F, Rhee KE, Lumeng JC, Dorrestein PC, Knight R, Kim JH, Song SJ, Swafford AD. Multiomic Analyses of Nascent Preterm Infant Microbiomes Differentiation Suggest Opportunities for Targeted Intervention. Adv Biol (Weinh) 2022; 6:e2101313. [PMID: 35652166 PMCID: PMC10321678 DOI: 10.1002/adbi.202101313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 04/01/2022] [Indexed: 01/28/2023]
Abstract
The first week after birth is a critical time for the establishment of microbial communities for infants. Preterm infants face unique environmental impacts on their newly acquired microbiomes, including increased incidence of cesarean section delivery and exposure to antibiotics as well as delayed enteral feeding and reduced human interaction during their intensive care unit stay. Using contextualized paired metabolomics and 16S sequencing data, the development of the gut, skin, and oral microbiomes of infants is profiled daily for the first week after birth, and it is found that the skin microbiome appears robust to early life perturbation, while direct exposure of infants to antibiotics, rather than presumed maternal transmission, delays microbiome development and prevents the early differentiation based on body site regardless of delivery mode. Metabolomic analyses identify the development of all gut metabolomes of preterm infants toward full-term infant profiles, but a significant increase of primary bile acid metabolism only in the non-antibiotic treated vaginally birthed late preterm infants. This study provides a framework for future multi-omic, multibody site analyses on these high-risk preterm infant populations and suggests opportunities for monitoring and intervention, with infant antibiotic exposure as the primary driver of delays in microbiome development.
Collapse
Affiliation(s)
- Stephanie B. Orchanian
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Present address: Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA
- These authors contributed equally to this work
| | - Julia M. Gauglitz
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
- Collaborative Mass Spectrometry Innovation Center, University of California San Diego, La Jolla, CA, USA
- These authors contributed equally to this work
| | - Stephen Wandro
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- These authors contributed equally to this work
| | - Kelly C. Weldon
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
- Collaborative Mass Spectrometry Innovation Center, University of California San Diego, La Jolla, CA, USA
| | - Megan Doty
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
- Present address: Neonatal Intensive Care Unit, Kapi’olani Medical Center for Women & Children, Honolulu, HI, USA
| | - Kristina Stillwell
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
- Present address: Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Shalisa Hansen
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
| | - Lingjing Jiang
- Division of Biostatistics, University of California San Diego, La Jolla, CA, USA
| | - Fernando Vargas
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
- Collaborative Mass Spectrometry Innovation Center, University of California San Diego, La Jolla, CA, USA
- Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
- Present address: Perinatal Institute, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kyung E. Rhee
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Julie C. Lumeng
- Department of Pediatrics, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Pieter C. Dorrestein
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
- Collaborative Mass Spectrometry Innovation Center, University of California San Diego, La Jolla, CA, USA
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Rob Knight
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA 92093, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Jae H. Kim
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
- Present address: Perinatal Institute, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Se Jin Song
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
| | - Austin D. Swafford
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
21
|
Zeng S, Ying J, Li S, Qu Y, Mu D, Wang S. First 1000 Days and Beyond After Birth: Gut Microbiota and Necrotizing Enterocolitis in Preterm Infants. Front Microbiol 2022; 13:905380. [PMID: 35801107 PMCID: PMC9253634 DOI: 10.3389/fmicb.2022.905380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/25/2022] [Indexed: 11/13/2022] Open
Abstract
Preterm birth remains a major maternal and infant health issue worldwide particularly with an increase in the global preterm birth rate, which requires more interventions to manage the consequences of preterm birth. In addition to traditional complications, recent studies have shown that the succession of gut microbiota of preterm infants is disordered due to the systemic physiological immaturity, which confers negative influences on the growth, development, and health of infants. In the present study, we briefly discussed the prevalence of preterm birth worldwide and then highlighted the signatures of gut microbiota in preterm infants within the first 1000 days of life after the birth categorized into birth, infancy, and childhood. Afterward, we focused on the potential association of clinical phenotypes typically associated with preterm birth (i.e., necrotizing enterocolitis) with gut microbiota, and the potential directions for future studies in this field are finally discussed.
Collapse
|
22
|
A Pilot Study on Donor Human Milk Microbiota: A Comparison with Preterm Human Milk Microbiota and the Effect of Pasteurization. Nutrients 2022; 14:nu14122483. [PMID: 35745213 PMCID: PMC9227689 DOI: 10.3390/nu14122483] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 02/04/2023] Open
Abstract
Human milk (HM) is the best feeding option for preterm infants; however, when mother’s own milk (MOM) is not available, pasteurized donor human milk (DHM) is the best alternative. In this study, we profiled DHM microbiota (19 samples) using 16S rRNA amplicon sequencing and compared its compositional features with the MOM microbiota (14 samples) from mothers who delivered prematurely (PT-MOM). As a secondary study aim, we assessed the specific effect of pasteurization on the characteristics of the DHM microbiota. DHM showed significantly higher alpha diversity and significant segregation from PT-MOM. Compositionally, the PT-MOM microbiota had a significantly higher proportion of Staphylococcus than DHM, with Streptococcus tending to be and Pseudomonas being significantly overrepresented in DHM compared with the PT-MOM samples. Furthermore, pasteurization affected the HM microbiota structure, with a trend towards greater biodiversity and some compositional differences following pasteurization. This pilot study provided further evidence on the HM microbial ecosystem, demonstrating that the DHM microbiota differs from the PT-MOM microbiota, possibly due to inherent differences between HM donors and mothers delivering prematurely, and that pasteurization per se impacts the HM microbiota. Knowledge about HM microbiota needs to be acquired by investigating the effect of DHM processing to develop strategies aimed at improving DHM quality while guaranteeing its microbiological safety.
Collapse
|
23
|
Rajoka MSR, Mehwish HM, Kitazawa H, Barba FJ, Berthelot L, Umair M, Zhu Q, He Z, Zhao L. Techno-functional properties and immunomodulatory potential of exopolysaccharide from Lactiplantibacillus plantarum MM89 isolated from human breast milk. Food Chem 2022; 377:131954. [PMID: 34973591 DOI: 10.1016/j.foodchem.2021.131954] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 12/23/2021] [Accepted: 12/23/2021] [Indexed: 11/18/2022]
Abstract
An exopolysaccharide, designated as MM89-EPS, was isolated from Lactiplantibacillus plantarum MM89. It was comprised of glucose and mannose molecules with an average molecular weight of 138 kDa. FTIR and NMR spectra showed that MM89-EPS had characteristic polysaccharide functional groups. MM89-EPS displayed excellent water solubility and capacities to retain water and oil due to its porous structure. MM89-EPS exhibited no significant cytotoxicity on RAW264.7 cells and showed strong immunomodulatory activity by increasing phagocytosis, acid phosphatase activity, and cytokine production in RAW264.7 cells. Furthermore, an in vivo study revealed that splenic indices, intestinal IgA levels, serum cytokine levels, and lymphocyte proliferation were increased in an MM89-EPS-treated cyclophosphamide-induced immunosuppressed mouse model. To summarize, our results indicate that MM89-EPS can efficiently enhance the immunostimulatory activity of immune cells and an immunosuppressed mouse model. Hence, MM89-EPS may be use as a potential source of immunomodulatory agent in various food products.
Collapse
Affiliation(s)
- Muhammad Shahid Riaz Rajoka
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, PR China; School of Pharmaceutical Science, Health Science Center, Shenzhen University, Shenzhen 518060, PR China; Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | - Hafiza Mahreen Mehwish
- School of Pharmaceutical Science, Health Science Center, Shenzhen University, Shenzhen 518060, PR China; Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | - Haruki Kitazawa
- Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan.
| | - Francisco J Barba
- Department of Preventive Medicine and Public Health, Food Science, Toxicology and Forensic Medicine, Faculty of Pharmacy, Universitat de València, Avda. Vicent Andrés Estellés, 46100 València, Spain.
| | - Laureline Berthelot
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France.
| | - Muhammad Umair
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, PR China.
| | - Qinchang Zhu
- School of Pharmaceutical Science, Health Science Center, Shenzhen University, Shenzhen 518060, PR China.
| | - Zhendan He
- School of Pharmaceutical Science, Health Science Center, Shenzhen University, Shenzhen 518060, PR China; College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, PR China.
| | - Liqing Zhao
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, PR China.
| |
Collapse
|
24
|
Aziz M, Prince JM, Wang P. Gut microbiome and necrotizing enterocolitis: Understanding the connection to find a cure. Cell Host Microbe 2022; 30:612-616. [PMID: 35550664 DOI: 10.1016/j.chom.2022.04.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Necrotizing enterocolitis (NEC), a cause of death among premature babies, has defied therapeutics for decades. Bacterial analyses have expanded insights into NEC pathophysiology and roles of the gut microbiome. We discuss the contribution of the gut microbiome and potential therapeutics, notably lactadherin, that may promote gut homeostasis to alleviate NEC.
Collapse
Affiliation(s)
- Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, USA; Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Jose M Prince
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, USA; Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA; Division of Pediatric Surgery, Cohen Children's Medical Center at Hofstra/Northwell, New Hyde Park, NY, USA.
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, USA; Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA.
| |
Collapse
|
25
|
Notarbartolo V, Giuffrè M, Montante C, Corsello G, Carta M. Composition of Human Breast Milk Microbiota and Its Role in Children's Health. Pediatr Gastroenterol Hepatol Nutr 2022; 25:194-210. [PMID: 35611376 PMCID: PMC9110848 DOI: 10.5223/pghn.2022.25.3.194] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 04/02/2022] [Indexed: 12/26/2022] Open
Abstract
Human milk contains a number of nutritional and bioactive molecules including microorganisms that constitute the so-called "Human Milk Microbiota (HMM)". Recent studies have shown that not only bacterial but also viral, fungal, and archaeal components are present in the HMM. Previous research has established, a "core" microbiome, consisting of Firmicutes (i.e., Streptococcus, Staphylococcus), Proteobacteria (i.e., Serratia, Pseudomonas, Ralstonia, Sphingomonas, Bradyrhizobium), and Actinobacteria (i.e., Propionibacterium, Corynebacterium). This review aims to summarize the main characteristics of HMM and the role it plays in shaping a child's health. We reviewed the most recent literature on the topic (2019-2021), using the PubMed database. The main sources of HMM origin were identified as the retrograde flow and the entero-mammary pathway. Several factors can influence its composition, such as maternal body mass index and diet, use of antibiotics, time and type of delivery, and mode of breastfeeding. The COVID-19 pandemic, by altering the mother-infant dyad and modifying many of our previous habits, has emerged as a new risk factor for the modification of HMM. HMM is an important contributor to gastrointestinal colonization in children and therefore, it is fundamental to avoid any form of perturbation in the HMM that can alter the microbial equilibrium, especially in the first 100 days of life. Microbial dysbiosis can be a trigger point for the development of necrotizing enterocolitis, especially in preterm infants, and for onset of chronic diseases, such as asthma and obesity, later in life.
Collapse
Affiliation(s)
- Veronica Notarbartolo
- Department of Health Promotion, Mather and Child Care, Internal Medicine and Medical Specialities, University of Palermo, Palermo, Italy
| | - Mario Giuffrè
- Department of Health Promotion, Mather and Child Care, Internal Medicine and Medical Specialities, University of Palermo, Palermo, Italy
| | - Claudio Montante
- Department of Health Promotion, Mather and Child Care, Internal Medicine and Medical Specialities, University of Palermo, Palermo, Italy
| | - Giovanni Corsello
- Department of Health Promotion, Mather and Child Care, Internal Medicine and Medical Specialities, University of Palermo, Palermo, Italy
| | - Maurizio Carta
- Department of Health Promotion, Mather and Child Care, Internal Medicine and Medical Specialities, University of Palermo, Palermo, Italy
| |
Collapse
|
26
|
Lyons KE, Shea CAO, Grimaud G, Ryan CA, Dempsey E, Kelly AL, Ross RP, Stanton C. The human milk microbiome aligns with lactation stage and not birth mode. Sci Rep 2022; 12:5598. [PMID: 35379843 PMCID: PMC8979980 DOI: 10.1038/s41598-022-09009-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 03/03/2022] [Indexed: 12/13/2022] Open
Abstract
We analysed the human milk microbiome in a cohort of 80 lactating women and followed the dynamics in taxa over the course of lactation from birth to 6 months. Two hundred and thirty one milk samples were collected from full-term lactating women at 1, 4, 8 and 24 weeks following birth and analysed for microbiota composition using 16S rRNA sequencing. A significant decrease in milk microbiota diversity was observed throughout the first 6 months of lactation, with the greatest difference seen between week 8 and week 24. Nine genera predominated in milk over lactation from week 1 to week 24, comprising of Staphylococcus, Streptococcus, Pseudomonas, Acinetobacter, Bifidobacterium, Mesorhizobium, Brevundimonas, Flavobacterium, and Rhodococcus; however, fluctuations in these core genera were apparent over time. There was a significant effect of stage of lactation on the microbiome, while no effect of birth mode, infant sex and maternal BMI was observed throughout lactation. Streptococcus had the highest mean relative abundance at week 1 and 24 (17.3% and 24% respectively), whereas Pseudomonas predominated at week 4 (22%) and week 8 (19%). Bifidobacterium and Lactobacillus had the highest mean relative abundance at week 4 (5% and 1.4% respectively), and occurred at a relative abundance of ≤ 1% at all other time points. A decrease in milk microbiota diversity throughout lactation was also observed. This study concluded that lactation stage was the primary driving factor in milk microbiota compositional changes over lactation from birth to 6 months, while mode of delivery was not a factor driving compositional changes throughout human lactation.
Collapse
|
27
|
de Weerth C, Aatsinki AK, Azad MB, Bartol FF, Bode L, Collado MC, Dettmer AM, Field CJ, Guilfoyle M, Hinde K, Korosi A, Lustermans H, Mohd Shukri NH, Moore SE, Pundir S, Rodriguez JM, Slupsky CM, Turner S, van Goudoever JB, Ziomkiewicz A, Beijers R. Human milk: From complex tailored nutrition to bioactive impact on child cognition and behavior. Crit Rev Food Sci Nutr 2022; 63:7945-7982. [PMID: 35352583 DOI: 10.1080/10408398.2022.2053058] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human milk is a highly complex liquid food tailor-made to match an infant's needs. Beyond documented positive effects of breastfeeding on infant and maternal health, there is increasing evidence that milk constituents also impact child neurodevelopment. Non-nutrient milk bioactives would contribute to the (long-term) development of child cognition and behavior, a process termed 'Lactocrine Programming'. In this review we discuss the current state of the field on human milk composition and its links with child cognitive and behavioral development. To promote state-of-the-art methodologies and designs that facilitate data pooling and meta-analytic endeavors, we present detailed recommendations and best practices for future studies. Finally, we determine important scientific gaps that need to be filled to advance the field, and discuss innovative directions for future research. Unveiling the mechanisms underlying the links between human milk and child cognition and behavior will deepen our understanding of the broad functions of this complex liquid food, as well as provide necessary information for designing future interventions.
Collapse
Affiliation(s)
- Carolina de Weerth
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, EN Nijmegen, The Netherlands
| | - Anna-Katariina Aatsinki
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
| | - Meghan B Azad
- Department of Pediatrics and Child Health, Manitoba Interdisciplinary Lactation Centre, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Frank F Bartol
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
| | - Lars Bode
- Department of Pediatrics and Mother-Milk-Infant Center of Research Excellence (MOMI CORE), University of California San Diego, La Jolla, California, USA
| | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Paterna, Valencia, Spain
| | - Amanda M Dettmer
- Yale Child Study Center, Yale School of Medicine, New Haven, Connecticut, USA
| | - Catherine J Field
- Department of Agricultural, Food and Nutritional Science, College of Basic and Applied Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Meagan Guilfoyle
- Department of Anthropology, Indiana University, Bloomington, Indiana, USA
| | - Katie Hinde
- School of Human Evolution and Social Change, Arizona State University, Tempe, Arizona, USA
| | - Aniko Korosi
- Swammerdam Institute for Life Sciences, Center for Neuroscience, Brain Plasticity group, University of Amsterdam, Amsterdam, The Netherlands
| | - Hellen Lustermans
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, EN Nijmegen, The Netherlands
| | - Nurul Husna Mohd Shukri
- Department of Nutrition, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| | - Sophie E Moore
- Department of Women & Children's Health, King's College London, St Thomas' Hospital, London, UK
- School of Hygiene and Tropical Medicine, Nutrition Theme, MRC Unit The Gambia and the London, Fajara, The GambiaBanjul
| | - Shikha Pundir
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Juan Miguel Rodriguez
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| | - Carolyn M Slupsky
- Department of Nutrition and Department of Food Science and Technology, University of California, Davis, California, USA
| | - Sarah Turner
- Department of Community Health Sciences, Manitoba Interdisciplinary Lactation Centre, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Johannes B van Goudoever
- Department of Pediatrics, Amsterdam UMC, University of Amsterdam, Vrije Universiteit, Emma Children's Hospital, Amsterdam, The Netherlands
| | - Anna Ziomkiewicz
- Department of Anthropology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Roseriet Beijers
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, EN Nijmegen, The Netherlands
- Department of Social Development, Behavioural Science Institute, Radboud University, Nijmegen, The Netherlands
| |
Collapse
|
28
|
Abstract
The neonatal body provides a range of potential habitats, such as the gut, for microbes. These sites eventually harbor microbial communities (microbiotas). A "complete" (adult) gut microbiota is not acquired by the neonate immediately after birth. Rather, the exclusive, milk-based nutrition of the infant encourages the assemblage of a gut microbiota of low diversity, usually dominated by bifidobacterial species. The maternal fecal microbiota is an important source of bacterial species that colonize the gut of infants, at least in the short-term. However, development of the microbiota is influenced by the use of human milk (breast feeding), infant formula, preterm delivery of infants, caesarean delivery, antibiotic administration, family details and other environmental factors. Following the introduction of weaning (complementary) foods, the gut microbiota develops in complexity due to the availability of a diversity of plant glycans in fruits and vegetables. These glycans provide growth substrates for the bacterial families (such as members of the Ruminococcaceae and Lachnospiraceae) that, in due course, will dominate the gut microbiota of the adult. Although current data are often fragmentary and observational, it can be concluded that the nutrition that a child receives in early life is likely to impinge not only on the development of the microbiota at that time but also on the subsequent lifelong, functional relationships between the microbiota and the human host. The purpose of this review, therefore, is to discuss the importance of promoting the assemblage of functionally robust gut microbiotas at appropriate times in early life.
Collapse
Affiliation(s)
- Gerald W. Tannock
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
29
|
Han SM, Derraik JGB, Binia A, Sprenger N, Vickers MH, Cutfield WS. Maternal and Infant Factors Influencing Human Milk Oligosaccharide Composition: Beyond Maternal Genetics. J Nutr 2021; 151:1383-1393. [PMID: 33768224 DOI: 10.1093/jn/nxab028] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 01/06/2021] [Accepted: 01/26/2021] [Indexed: 02/06/2023] Open
Abstract
Maternal genetics is a key determinant of human milk oligosaccharide (HMO) composition in human milk. Beyond genetic status, other factors influencing the HMO profile are poorly defined. Thus, we aimed to review the existing evidence on the associations between nongenetic maternal and infant factors and HMO composition. A systematic search was performed on PubMed and Web of Science (without a time restriction) to identify any relevant studies published. In total, 1056 results were obtained, of which 29 articles were selected to be included in this review. The range of factors investigated include lactation stage, maternal pre-pregnancy BMI (ppBMI), maternal age, parity, maternal diet, mode of delivery, infant gestational age, and infant sex. The data suggest that, beyond maternal genetics, HMO composition seems to be influenced by all these factors, but the underlining mechanisms remain speculative. The published evidence is discussed in this review, along with potential implications for infant growth and development. For example, 2'-fucosyllactose, which was reportedly increased in mothers with higher ppBMIs, was also associated with increased infant weight and height. In addition, greater levels of sialylated HMOs after preterm birth may support brain development in these infants.
Collapse
Affiliation(s)
- Soo Min Han
- Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - José G B Derraik
- Liggins Institute, The University of Auckland, Auckland, New Zealand.,Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden.,Department of Endocrinology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Aristea Binia
- Nestlé Research, Société des Produits Nestlé SA, Lausanne, Switzerland
| | - Norbert Sprenger
- Nestlé Research, Société des Produits Nestlé SA, Lausanne, Switzerland
| | - Mark H Vickers
- Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Wayne S Cutfield
- Liggins Institute, The University of Auckland, Auckland, New Zealand.,A Better Start-National Science Challenge, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
30
|
Chen T, Qin Y, Chen M, Zhang Y, Wang X, Dong T, Chen G, Sun X, Lu T, White RA, Ye P, Tun HM, Xia Y. Gestational diabetes mellitus is associated with the neonatal gut microbiota and metabolome. BMC Med 2021; 19:120. [PMID: 34039350 PMCID: PMC8157751 DOI: 10.1186/s12916-021-01991-w] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 04/23/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Gestational diabetes mellitus (GDM) is a metabolic disease that occurs in pregnant women and increases the risk for the development of diabetes. The relationship between GDM and meconium microbiota and metabolome remains incompletely understood. METHODS Four hundred eighteen mothers (147 women with GDM and 271 normal pregnant women) and their neonates from the GDM Mother and Child Study were included in this study. Meconium microbiota were profiled by 16S rRNA gene sequencing. Meconium and maternal serum metabolome were examined by UPLC-QE. RESULTS Microbial communities in meconium were significantly altered in neonates from the GDM mothers. A reduction in alpha diversity was observed in neonates of GDM mothers. At the phylum level, the abundance of Firmicutes and Proteobacteria changed significantly in neonates of GDM mothers. Metabolomic analysis of meconium showed that metabolic pathways including taurine and hypotaurine metabolism, pyrimidine metabolism, beta-alanine metabolism, and bile acid biosynthesis were altered in GDM subjects. Several changed metabolites varying by the similar trend across the maternal serum and neonatal meconium were observed. CONCLUSION Altogether, these findings suggest that GDM could alter the serum metabolome and is associated with the neonatal meconium microbiota and metabolome, highlighting the importance of maternal factors on early-life metabolism.
Collapse
Affiliation(s)
- Ting Chen
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China
| | - Yufeng Qin
- Department of Microbes and Infection, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Minjian Chen
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yuqing Zhang
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Xu Wang
- Department of Endocrinology, Children's Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - Tianyu Dong
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Guanglin Chen
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - Xian Sun
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Ting Lu
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Richard Allen White
- Department of Bioinformatics and Genomics at University of North Carolina, Charlotte, USA.,RAW Molecular Systems LLC, Concord, USA
| | - Peng Ye
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Hein M Tun
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Yankai Xia
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, 211166, China. .,Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
31
|
Ruiz L, Alba C, García-Carral C, Jiménez EA, Lackey KA, McGuire MK, Meehan CL, Foster J, Sellen DW, Kamau-Mbuthia EW, Kamundia EW, Mbugua S, Moore SE, Prentice AM, Gindola K D, Otoo GE, Pareja RG, Bode L, McGuire MA, Williams JE, Rodríguez JM. Comparison of Two Approaches for the Metataxonomic Analysis of the Human Milk Microbiome. Front Cell Infect Microbiol 2021; 11:622550. [PMID: 33842385 PMCID: PMC8027255 DOI: 10.3389/fcimb.2021.622550] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 03/05/2021] [Indexed: 01/04/2023] Open
Abstract
Recent work has demonstrated the existence of large inter-individual and inter-population variability in the microbiota of human milk from healthy women living across variable geographical and socio-cultural settings. However, no studies have evaluated the impact that variable sequencing approaches targeting different 16S rRNA variable regions may have on the human milk microbiota profiling results. This hampers our ability to make meaningful comparisons across studies. In this context, the main purpose of the present study was to re-process and re-sequence the microbiome in a large set of human milk samples (n = 412) collected from healthy women living at diverse international sites (Spain, Sweden, Peru, United States, Ethiopia, Gambia, Ghana and Kenya), by targeting a different 16S rRNA variable region and reaching a larger sequencing depth. Despite some differences between the results obtained from both sequencing approaches were notable (especially regarding alpha and beta diversities and Proteobacteria representation), results indicate that both sequencing approaches revealed a relatively consistent microbiota configurations in the studied cohorts. Our data expand upon the milk microbiota results we previously reported from the INSPIRE cohort and provide, for the first time across globally diverse populations, evidence of the impact that different DNA processing and sequencing approaches have on the microbiota profiles obtained for human milk samples. Overall, our results corroborate some similarities regarding the microbial communities previously reported for the INSPIRE cohort, but some differences were also detected. Understanding the impact of different sequencing approaches on human milk microbiota profiles is essential to enable meaningful comparisons across studies. Clinical Trial Registration www.clinicaltrials.gov, identifier NCT02670278.
Collapse
Affiliation(s)
- Lorena Ruiz
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
- *Correspondence: Lorena Ruiz, ; Juan Miguel Rodriguez,
| | - Claudio Alba
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| | - Cristina García-Carral
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| | - Esther A. Jiménez
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| | - Kimberly A. Lackey
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID, United States
| | - Michelle K. McGuire
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID, United States
| | - Courtney L. Meehan
- Department of Anthropology, Washington State University, Pullman, WA, United States
| | - James Foster
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID, United States
| | - Daniel W. Sellen
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | | | | | - Samwel Mbugua
- Department of Human Nutrition, Egerton University, Nakuru, Kenya
| | - Sophie E. Moore
- Division of Women’s Health, King’s College London, London, United Kingdom
- MRC Unit, Serekunda, Gambia
| | - Andrew M. Prentice
- MRC International Nutrition Group, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Debela Gindola K
- Department of Anthropology, Hawassa University, Hawassa, Ethiopia
| | - Gloria E. Otoo
- Department of Nutrition and Food Science, University of Ghana, Accra, Ghana
| | | | - Lars Bode
- Department of Pediatrics and Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (MOMI CoRE), University of California, San Diego, La Jolla, CA, United States
| | - Mark A. McGuire
- Department of Animal and Veterinary Science, University of Idaho, Moscow, ID, United States
| | - Janet E. Williams
- Department of Animal and Veterinary Science, University of Idaho, Moscow, ID, United States
| | - Juan M. Rodríguez
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
- *Correspondence: Lorena Ruiz, ; Juan Miguel Rodriguez,
| |
Collapse
|
32
|
Sánchez Luna M, Martin SC, Gómez-de-Orgaz CS. Human milk bank and personalized nutrition in the NICU: a narrative review. Eur J Pediatr 2021; 180:1327-1333. [PMID: 33244710 PMCID: PMC7691070 DOI: 10.1007/s00431-020-03887-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/14/2020] [Accepted: 11/20/2020] [Indexed: 01/01/2023]
Abstract
The number of infants born preterm including extremely premature babies is rising worldwide, particularly in low- and middle-income countries, which challenge neonatologists and milk banks for the provision of the most adequate nutrition for successful infant's growth and development. The benefits of mother's own milk (MOM) have been extensively recognized, but the use of donor milk (DM) is a commonly routine practice in preterm neonates admitted to the NICU. Pasteurized mature milk from milk banks is not the same composition than the mother's colostrum and premature milk, the characteristics of which protect the infant from the risk for necrotizing enterocolitis, late-onset sepsis, and other comorbidities associated with prematurity. The development of a personalized nutrition unit (PNU) allows to obtain DM from mothers who have their infants admitted to the NICU and produce an excess of milk, a practice that matches MOM by gestational age and the stage of lactation, ensuring an adequate composition of DM to target the nutritional requirements of premature infants.Conclusion: This narrative review presents salient data of our current knowledge and concerns regarding milk feeding of preterm infants in the NICU, with special emphasis on personalized DM as a result of establishing a PNU. What is Known: • Donor milk bank is mature or pooled milk from lactating mothers at different stages of lactation. • Milk composition varies by gestational age and stage of lactation. What is New: • Donor milk from mothers delivered prematurely have the most adequate composition for preterm infant feeding. • Personalized nutrition for premature infants with preterm donor milk is feasible.
Collapse
Affiliation(s)
- Manuel Sánchez Luna
- Neonatology Department, Hospital General Universitario Gregorio Marañón, Universidad Complutense, C/ O'Donnell 48, E-28009, Madrid, Spain.
| | - Sylvia Caballero Martin
- grid.4795.f0000 0001 2157 7667Neonatology Department, Hospital General Universitario Gregorio Marañón, Universidad Complutense, C/ O’Donnell 48, E-28009 Madrid, Spain
| | - Carmen Sánchez Gómez-de-Orgaz
- grid.4795.f0000 0001 2157 7667Neonatology Department, Hospital General Universitario Gregorio Marañón, Universidad Complutense, C/ O’Donnell 48, E-28009 Madrid, Spain
| |
Collapse
|
33
|
Han SM, Binia A, Godfrey KM, El-Heis S, Cutfield WS. Do Human Milk Oligosaccharides Protect Against Infant Atopic Disorders and Food Allergy? Nutrients 2020; 12:nu12103212. [PMID: 33096669 PMCID: PMC7589050 DOI: 10.3390/nu12103212] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/14/2020] [Accepted: 10/20/2020] [Indexed: 12/28/2022] Open
Abstract
Atopic disorders (AD), often coexistent with food allergy (FA), start developing in early life and have lifelong health consequences. Breastfeeding is thought to be protective against AD and FA, but the data are controversial, and mechanisms are not well understood. Human milk oligosaccharides (HMOs) are complex carbohydrates that are abundant in human milk. These are thought to contribute to the development of the infant immune system by (i) promoting healthy microbiome, (ii) inhibiting pathogen binding to gut mucosa and (iii) modulating the immune system. Differences in microbiome composition between allergic and healthy infants have been observed, regardless of breastfeeding history. To date, limited studies have examined the preventive effects of HMOs on AD and FA in infants and current data relies on observation studies as trials of varying HMO intake through randomising individuals to breastfeeding are unethical. There is evidence for beneficial effects of breastfeeding on lowering the risks of FA, eczema and asthma but there are inconsistencies amongst studies in the duration of breastfeeding, diagnostic criteria for AD and the age at which the outcome was assessed. Furthermore, current analytical methods primarily used today only allow detection of 16-20 major HMOs while more than 100 types have been identified. More large-scale longitudinal studies are required to investigate the role of HMO composition and the impact of changes over the lactation period in preventing AD and FA later in life.
Collapse
Affiliation(s)
- Soo Min Han
- Liggins Institute, The University of Auckland, Auckland 1023, New Zealand;
| | - Aristea Binia
- Nestlé Research, Société des Produits Nestlé SA, 1000 Lausanne, Switzerland;
| | - Keith M. Godfrey
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton SO17 1BJ, UK;
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton SO17 1BJ, UK;
| | - Sarah El-Heis
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton SO17 1BJ, UK;
| | - Wayne S. Cutfield
- Liggins Institute, The University of Auckland, Auckland 1023, New Zealand;
- A Better Start—National Science Challenge, Liggins Institute, The University of Auckland, Auckland 1023, New Zealand
- Correspondence: ; Tel.: +64-9-923-4476
| |
Collapse
|