1
|
Picáns-Leis R, Vázquez-Mosquera ME, Pereira-Hernández M, Vizoso-González M, López-Valverde L, Barbosa-Gouveia S, López-Suárez O, López-Sanguos C, Bravo SB, García-González MA, Couce ML. Characterization of the functional component in human milk and identification of the molecular mechanisms undergoing prematurity. Clin Nutr 2025; 44:178-192. [PMID: 39700709 DOI: 10.1016/j.clnu.2024.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 11/06/2024] [Accepted: 12/09/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND AND AIMS Human milk (HM) is the earliest form of extrauterine communication between mother and infant, that could promote early programming. The aim of this study is to look for specific biological processes, particularly those undergoing prematurity, modulated by proteins and miRNAs of HM that could be implicated in growth and development. METHODS This is a prospective, observational, single center study in which we collected 48 human milk (HM) samples at two distinct stages of lactation: colostrum (first 72-96 h) and mature milk (at week 4 post-delivery) from mothers of very preterm newborns (<32 weeks) and term (≥37 and < 42 weeks). Qualitative and quantitative proteomic and transcriptomic analysis was done in our samples. RESULTS We performed isolation and characterization of HM extracellular vesicles (EVs) to carry out proteomic and transcriptomic analysis in colostrum (CM) and mature milk (MM). Proteomic analysis revealed a functional role of CM in immunological protection and MM in metabolic processes. TENA, TSP1 and OLF4, proteins with roles in immune response and inflammatory modulation, were upregulated in CM vs MM, particularly in preterm. HM modulation differed depending on gestational age (GA). The miRNAs identified in HM are implicated in structural functions, including growth and neurological development. miRNA-451a was differentially expressed between groups, and downregulated in preterm CM. CONCLUSIONS Because the particularities of each GA are reflected in the EVs content of HM, providing newborns with HM from their own mother is the optimal way for satisfying their specific needs. Although the role of the proteomic profile of CM and MM of different GA in relation to neurodevelopment has been previously described, this is the first study to show a complete functional characterization of HM (proteome, miRNA at the same time), unmasking the molecular mechanisms related to EVs signaling and their functional role in preterm.
Collapse
Affiliation(s)
- Rosaura Picáns-Leis
- Neonatology Department, Metabolic Unit, RICORS-SAMID, CIBERER, University Clinical Hospital of Santiago de Compostela, Spain; Health Research Institute of Santiago de Compostela (IDIS), Spain.
| | - María E Vázquez-Mosquera
- Neonatology Department, Metabolic Unit, RICORS-SAMID, CIBERER, University Clinical Hospital of Santiago de Compostela, Spain; Health Research Institute of Santiago de Compostela (IDIS), Spain.
| | - María Pereira-Hernández
- Health Research Institute of Santiago de Compostela (IDIS), Spain; Group of Genetics and Developmental Biology of Renal Diseases, Nephrology Laboratory, University Clinical Hospital of Santiago de Compostela, Spain; RICORS2040 (Kidney Disease), Santiago de Compostela, Spain.
| | - Marta Vizoso-González
- Health Research Institute of Santiago de Compostela (IDIS), Spain; Group of Genetics and Developmental Biology of Renal Diseases, Nephrology Laboratory, University Clinical Hospital of Santiago de Compostela, Spain.
| | - Laura López-Valverde
- Neonatology Department, Metabolic Unit, RICORS-SAMID, CIBERER, University Clinical Hospital of Santiago de Compostela, Spain; Health Research Institute of Santiago de Compostela (IDIS), Spain.
| | - Sofía Barbosa-Gouveia
- Neonatology Department, Metabolic Unit, RICORS-SAMID, CIBERER, University Clinical Hospital of Santiago de Compostela, Spain; Health Research Institute of Santiago de Compostela (IDIS), Spain.
| | - Olalla López-Suárez
- Neonatology Department, Metabolic Unit, RICORS-SAMID, CIBERER, University Clinical Hospital of Santiago de Compostela, Spain; Health Research Institute of Santiago de Compostela (IDIS), Spain.
| | - Carolina López-Sanguos
- Neonatology Department, Metabolic Unit, RICORS-SAMID, CIBERER, University Clinical Hospital of Santiago de Compostela, Spain; Health Research Institute of Santiago de Compostela (IDIS), Spain.
| | - Susana B Bravo
- Health Research Institute of Santiago de Compostela (IDIS), Spain; Proteomic Platform, University Clinical Hospital of Santiago de Compostela, Spain.
| | - Miguel A García-González
- Health Research Institute of Santiago de Compostela (IDIS), Spain; Group of Genetics and Developmental Biology of Renal Diseases, Nephrology Laboratory, University Clinical Hospital of Santiago de Compostela, Spain; RICORS2040 (Kidney Disease), Santiago de Compostela, Spain.
| | - María L Couce
- Neonatology Department, Metabolic Unit, RICORS-SAMID, CIBERER, University Clinical Hospital of Santiago de Compostela, Spain; Health Research Institute of Santiago de Compostela (IDIS), Spain.
| |
Collapse
|
2
|
Yazdanbakhsh M, De Andrade V, Spiesser-Robelet L, Gagnayre R. The need for educational intervention for breastfeeding women and the professional practice of midwives in France to promote breastfeeding: A joint explanatory study. Eur J Midwifery 2024; 8:EJM-8-73. [PMID: 39664091 PMCID: PMC11633045 DOI: 10.18332/ejm/191176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 04/22/2024] [Accepted: 07/13/2024] [Indexed: 12/13/2024] Open
Abstract
In France, breastfeeding prevalence is high at birth, but its continuation remains low compared with other high-income countries, despite worldwide public health recommendations. Midwives offer parenting classes in an experimental manner without considering the importance of education in their interventions. The objectives of this study were to identify the teaching strategies and learning environments offered by midwives and their effect on women's perception of usefulness and their breastfeeding competence, to assess midwives' perception of usefulness and their pedagogical competencies. A comparative mixed study of 20 hospital midwives and 53 breastfeeding women (at 3 and 30 days postpartum) was conducted between January and August 2022 in two maternity units in France. Comparing the two periods, positive effects were found about breastfeeding women's level of knowledge: usefulness of learning theoretical (p=0.01) and practical (p=0.00) knowledge; and their breastfeeding management: signs of lactation (p=0.00), breast engorgement (p=0.04), and behavior (p=0.04). It positively reinforced the development of self-esteem (p=0.00) and commitment to breastfeeding (p=0.00). Midwives expressed strong motivation to use an appropriate teaching strategy and provide a supportive learning environment for women to improve their educational interventions (mean motivation score 7.7/10). The study results can promote research to examine educational interventions modeled according to the theories in education. Critical realism can be used to evaluate these interventions to elucidate how a program based on educational engineering can contribute to breastfeeding promotion and achieving the 2030 goals of WHO. CLINICAL TRIAL REGISTRATION The study was registered on the official website of ClinicalTrials.gov. IDENTIFIER ID NCT05271812.
Collapse
Affiliation(s)
- Mehrnoosh Yazdanbakhsh
- Educations and Health Promotion Laboratory, Sorbonne Paris Nord University, Villetaneuse, France
| | - Vincent De Andrade
- Educations and Health Promotion Laboratory, Sorbonne Paris Nord University, Villetaneuse, France
| | | | - Rémi Gagnayre
- Educations and Health Promotion Laboratory, Sorbonne Paris Nord University, Villetaneuse, France
| |
Collapse
|
3
|
Wijenayake S, Eisha S, Purohit MK, McGowan PO. Milk derived extracellular vesicle uptake in human microglia regulates the DNA methylation machinery : Short title: milk-derived extracellular vesicles and the epigenetic machinery. Sci Rep 2024; 14:28630. [PMID: 39562680 PMCID: PMC11576889 DOI: 10.1038/s41598-024-79724-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 11/12/2024] [Indexed: 11/21/2024] Open
Abstract
Mammalian milk contains milk-derived extracellular vesicles (MEVs), a group of biological nanovesicles that transport macromolecules. Their ability to cross the blood brain barrier and the presence of cargo capable of modifying gene function have led to the hypothesis that MEVs may play a role in brain function and development. Here, we investigated the uptake of MEVs by human microglia cells in vitro and explored the functional outcomes of MEV uptake. We examined the expression of the miR-148/152 family, highly abundant MEV microRNAs, that directly suppress the translation of DNA methyltransferase (DNMT) enzymes crucial for catalyzing DNA methylation modifications. We also measured phenotypic and inflammatory gene expression in baseline homeostatic and IFN-γ primed microglia to determine if MEVs induce anti-inflammatory effects. We found that MEVs are taken up and localize in baseline and primed microglia. In baseline microglia, MEV supplementation reduced miR-148a-5P levels, increased DNMT1 transcript, protein abundance, and enzymatic activity, compared to cells that did not receive MEVs. In primed microglia, MEV supplementation decreased miR-148a-5P levels and increased DNMT1 protein abundance, but DNMT1 transcript and enzymatic levels remained unchanged. Contrary to predictions, MEV supplementation failed to attenuate pro-inflammatory IL1β expression in primed microglia. This study provides the first evidence of MEV uptake by a brain macrophage, suggesting a potential role in regulating epigenetic machinery and neuroimmune modulation.
Collapse
Affiliation(s)
- Sanoji Wijenayake
- Department of Biology, The University of Winnipeg, Winnipeg, Manitoba, Canada.
- Department of Biological Sciences and Center for Environmental Epigenetics and Development, Scarborough Campus, University of Toronto, Toronto, ON, Canada.
| | - Shafinaz Eisha
- Department of Biological Sciences and Center for Environmental Epigenetics and Development, Scarborough Campus, University of Toronto, Toronto, ON, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Mansi Kamlesh Purohit
- Department of Biological Sciences and Center for Environmental Epigenetics and Development, Scarborough Campus, University of Toronto, Toronto, ON, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Patrick Owen McGowan
- Department of Biological Sciences and Center for Environmental Epigenetics and Development, Scarborough Campus, University of Toronto, Toronto, ON, Canada.
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada.
- Department of Psychology, University of Toronto, Toronto, ON, Canada.
- Department of Physiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
4
|
Graspeuntner S, Lupatsii M, van Zandbergen V, Dammann MT, Pagel J, Nguyen DN, Humberg A, Göpel W, Herting E, Rupp J, Härtel C, Fortmann I. Infants < 90 days of age with late-onset sepsis display disturbances of the microbiome-immunity interplay. Infection 2024:10.1007/s15010-024-02396-6. [PMID: 39541036 DOI: 10.1007/s15010-024-02396-6] [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: 01/23/2024] [Accepted: 09/09/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVE We hypothesized that previously healthy infants < 90 days of age with late-onset sepsis (LOS) have disturbances of the gut microbiome with yet undefined specific immunological patterns. METHODS We performed a prospective single-center convenience sample study between January 2019 and July 2021 in a case-control design. Routine diagnostics included conventional cultures (blood, cerebrospinal fluid, urine), PCRs and inflammatory markers in infants aged < 90 days with clinical LOS. We additionally analyzed blood lymphocyte subsets including CD4 + CD25 + forkhead box protein (FoxP3)+ Tregs and performed 16 S rRNA sequencing of stool samples, both compared to age-matched healthy controls. Results were adjusted for potential confounders that may influence microbial composition. RESULTS 51 infants with fever and clinical LOS were enrolled. Bacterial sepsis was diagnosed in n = 24 (47.1%) and viral infection in n = 13 (25.5%) infants, whereas in 14 (27.3%) infants the cause of fever remained undetermined. When compared to healthy controls, the gut microbiome of LOS infants at disease onset was characterized by a shift in community composition, specifically, decreased abundance of B. longum and an increase of Bacteroidia spp. Intriguingly, the abundance of B. longum negatively correlated with the frequency of blood CD4-positive cells in healthy controls but not in infants with LOS. At one year of age, we observed microbiome differences in infants with history of LOS when compared to healthy controls, such as an increased gut microbial diversity. CONCLUSION Our data suggest potential signatures of the microbiome-immunity interplay in infants with LOS, which should be investigated further as possible targets for prevention.
Collapse
Affiliation(s)
- Simon Graspeuntner
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Lübeck, Germany
| | - Mariia Lupatsii
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Vera van Zandbergen
- Department of Pediatrics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Marie-Theres Dammann
- Department of Pediatrics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Julia Pagel
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Lübeck, Germany
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Duc Ninh Nguyen
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Wolfgang Göpel
- Department of Pediatrics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Egbert Herting
- Department of Pediatrics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Jan Rupp
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Lübeck, Germany
| | - Christoph Härtel
- Department of Pediatrics, University Hospital of Würzburg, Würzburg, Germany
| | - Ingmar Fortmann
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Lübeck, Germany.
- Department of Pediatrics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany.
| |
Collapse
|
5
|
Badr L, Yasir M, Alkhaldy AA, Soliman SA, Ganash M, Turkistani SA, Jiman-Fatani AA, Al-Zahrani IA, Azhar EI. Genomic evaluation of the probiotic and pathogenic features of Enterococcus faecalis from human breast milk and comparison with the isolates from animal milk and clinical specimens. PeerJ 2024; 12:e18392. [PMID: 39494274 PMCID: PMC11529597 DOI: 10.7717/peerj.18392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 10/02/2024] [Indexed: 11/05/2024] Open
Abstract
Enterococcus faecalis is considered a probiotic, commensal lactic acid bacterium in human breast milk (HBM), but there are circulating antibiotic resistant and virulence determinants that could pose a risk in some strains. The study aimed to conduct genomic analysis of E. faecalis isolates recovered from HBM and animal milk and to evaluate their probiotic and pathogenic features through comparative genomics with isolates from clinical specimens (e.g., urine, wound, and blood). Genomic analysis of 61 isolates was performed, including E. faecalis isolates recovered from HBM in Saudi Arabia. Genome sequencing was conducted using the MiSeq system. The fewest antibiotic resistance genes (lsaA, tetM, ermB) were identified in isolates from HBM and animal milk compared to clinical isolates. Several known and unknown mutations in the gyrA and parC genes were observed in clinical isolates. However, 11 virulence genes were commonly found in more than 95% of isolates, and 13 virulence genes were consistently present in the HBM isolates. Phylogenetically, the HBM isolates from China clustered with the probiotic reference strain Symbioflor 1, but all isolates from HBM and animal milk clustered separately from the clinical reference strain V583. Subsystem functions 188 of 263 were common in all analyzed genome assemblies. Regardless of the source of isolation, genes associated with carbohydrate metabolism, fatty acid, and vitamin biosynthesis were commonly found in E. faecalis isolates. In conclusion, comparative genomic analysis can help distinguish the probiotic or pathogenic potential of E. faecalis based on genomic features.
Collapse
Affiliation(s)
- Lobna Badr
- Department of Biology, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Muhammad Yasir
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Areej A. Alkhaldy
- Department of Clinical Nutrition, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Samah A. Soliman
- Department of Nursing, Dr. Soliman Fakeeh Hospital, Jeddah, Saudi Arabia
| | - Magdah Ganash
- Department of Biology, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Safaa A. Turkistani
- Medical Laboratory Sciences, Fakeeh College for Medical Sciences, Jeddah, Saudi Arabia
| | - Asif A. Jiman-Fatani
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ibrahim A. Al-Zahrani
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Esam I. Azhar
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
6
|
Daniels VC, Monaco MH, Hirvonen J, Ouwehand AC, Jensen HM, Mukerjea R, Christensen N, Lehtinen MJ, Dilger RN, Donovan SM. Interactions between the human milk oligosaccharide 2'-fucosyllactose and Bifidobacterium longum subspecies infantis in influencing systemic immune development and function in piglets. Front Nutr 2024; 11:1444594. [PMID: 39525504 PMCID: PMC11543533 DOI: 10.3389/fnut.2024.1444594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction The oligosaccharide 2'-fucosyllactose (2'-FL) is a predominant component of human milk, serving as a prebiotic for gut microbiota and influencing immune development in infants. Bifidobacterium longum subspecies infantis (B. infantis) is a commensal bacterium found in breastfed infants. Both 2'-FL and a specific strain of B. infantis, Bi-26™, are commercially available. This study investigates the potential synbiotic relationship between 2'-FL and Bi-26™ on immune development. Methods Two-day-old piglets (n = 53) were randomized in a 2 × 2 design, receiving either a commercial milk replacer ad libitum without (CON) or with 1.0 g/L 2'-FL (FL). Piglets in each diet were further randomized to receive either glycerol stock alone or Bi-26™ (109 CFU) (BI and FLBI) orally once daily. On postnatal day (PND) 34/35, animals were euthanized, and blood was collected for serum cytokine analysis. Additionally, peripheral blood mononuclear cells (PBMCs) were isolated for ex vivo stimulation and flow cytometry analysis. Serum and ex vivo cytokines were analyzed using a multivariate model. All other outcomes were analyzed using a two-way ANOVA, considering prebiotic and probiotic fixed effects. The significance level was set at a p value <0.05, with trends reported for 0.05 < p < 0.1. Results Immune cell populations in PBMCs were unaffected by the experimental treatment. However, serum interleukin (IL)-1RA, IL-1β, IL-12, and IL-18 were all higher (p < 0.05) in the FL group than in the CON group. In isolated PBMCs, lipopolysaccharide (LPS) stimulation resulted in higher IL-1RA and a trend for higher IFN-γ secretion in the FL group vs. the CON group. Conclusion 2'-FL stimulates a balanced cytokine profile in healthy piglets without changing immune cell populations. When immune cells are stimulated ex vivo with LPS, 2'-FL primes T-cells for a proinflammatory response, which is moderated by co-administration of Bi-26™.
Collapse
Affiliation(s)
- Victoria C. Daniels
- Division of Nutritional Sciences, University of Illinois, Urbana, IL, United States
| | - Marcia H. Monaco
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL, United States
| | | | | | | | - Ratna Mukerjea
- IFF Health and Biosciences, Saint Louis, MO, United States
| | | | | | - Ryan N. Dilger
- Division of Nutritional Sciences, University of Illinois, Urbana, IL, United States
- Department of Animal Sciences, University of Illinois, Urbana, IL, United States
| | - Sharon M. Donovan
- Division of Nutritional Sciences, University of Illinois, Urbana, IL, United States
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL, United States
| |
Collapse
|
7
|
Bianco I, Ferrara C, Romano F, Loperfido F, Sottotetti F, El Masri D, Vincenti A, Cena H, De Giuseppe R. The Influence of Maternal Lifestyle Factors on Human Breast Milk Microbial Composition: A Narrative Review. Biomedicines 2024; 12:2423. [PMID: 39594990 PMCID: PMC11592219 DOI: 10.3390/biomedicines12112423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 11/28/2024] Open
Abstract
Human breast milk (HBM) is considered the gold standard for infant nutrition due to its optimal nutrient profile and complex composition of cellular and non-cellular components. Breastfeeding positively influences the newborn's gut microbiota and health, reducing the risk of conditions like gastrointestinal infections and chronic diseases (e.g., allergies, asthma, diabetes, and obesity). Research has revealed that HBM contains beneficial microbes that aid gut microbiota maturation through mechanisms like antimicrobial production and pathogen exclusion. The HBM microbiota composition can be affected by several factors, including gestational age, delivery mode, medical treatments, lactation stage, as well as maternal lifestyle habits (e.g., diet, physical activity, sleep quality, smoking, alcohol consumption, stress level). Particularly, lifestyle factors can play a significant role in shaping the HBM microbiota by directly modulating the microbial composition or influencing the maternal gut microbiota and influencing the HBM microbes through the enteromammary pathway. This narrative review of current findings summarized how maternal lifestyle influences HBM microbiota. While the influence of maternal diet on HBM microbiota is well-documented, indicating that dietary patterns, especially those rich in plant-based proteins and complex carbohydrates, can positively influence HBM microbiota, the impact of other lifestyle factors is poorly investigated. Maintaining a healthy lifestyle during pregnancy and breastfeeding is crucial for the health of both mother and baby. Understanding how maternal lifestyle factors influence microbial colonization of HBM, along with their interactions and impact, is key to developing new strategies that support the beneficial maturation of the infant's gut microbiota.
Collapse
Affiliation(s)
- Irene Bianco
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy; (I.B.); (C.F.); (F.R.); (F.L.); (D.E.M.); (A.V.); (H.C.); (R.D.G.)
| | - Chiara Ferrara
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy; (I.B.); (C.F.); (F.R.); (F.L.); (D.E.M.); (A.V.); (H.C.); (R.D.G.)
| | - Francesca Romano
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy; (I.B.); (C.F.); (F.R.); (F.L.); (D.E.M.); (A.V.); (H.C.); (R.D.G.)
| | - Federica Loperfido
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy; (I.B.); (C.F.); (F.R.); (F.L.); (D.E.M.); (A.V.); (H.C.); (R.D.G.)
| | - Francesca Sottotetti
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy; (I.B.); (C.F.); (F.R.); (F.L.); (D.E.M.); (A.V.); (H.C.); (R.D.G.)
| | - Dana El Masri
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy; (I.B.); (C.F.); (F.R.); (F.L.); (D.E.M.); (A.V.); (H.C.); (R.D.G.)
| | - Alessandra Vincenti
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy; (I.B.); (C.F.); (F.R.); (F.L.); (D.E.M.); (A.V.); (H.C.); (R.D.G.)
| | - Hellas Cena
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy; (I.B.); (C.F.); (F.R.); (F.L.); (D.E.M.); (A.V.); (H.C.); (R.D.G.)
- Clinical Nutrition Unit, General Medicine, Istituti Clinici Scientifici (ICS) Maugeri, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 27100 Pavia, Italy
| | - Rachele De Giuseppe
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy; (I.B.); (C.F.); (F.R.); (F.L.); (D.E.M.); (A.V.); (H.C.); (R.D.G.)
| |
Collapse
|
8
|
Çelik E, Cemali Ö, Şahin TÖ, Deveci G, Biçer NÇ, Hirfanoğlu İM, Ağagündüz D, Budán F. Human Breast Milk Exosomes: Affecting Factors, Their Possible Health Outcomes, and Future Directions in Dietetics. Nutrients 2024; 16:3519. [PMID: 39458514 PMCID: PMC11510026 DOI: 10.3390/nu16203519] [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: 09/21/2024] [Revised: 10/12/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Human breast milk is a complex biological fluid containing multifaceted biological compounds that boost immune and metabolic system development that support the short- and long-term health of newborns. Recent literature suggests that human breast milk is a substantial source of nutrients, bioactive molecules, and exosomes. Objectives: This review examines the factors influencing exosomes noted in human milk and the impacts of exosomes on infant health. Furthermore, it discusses potential future prospects for exosome research in dietetics. Methods: Through a narrative review of the existing literature, we focused on exosomes in breast milk, exosome components and their potential impact on exosome health. Results: Exosomes are single-membrane extracellular vesicles of endosomal origin, with an approximate radius of 20-200 nm. They are natural messengers that cells secrete to transport a wide range of diverse cargoes, including deoxyribonucleic acid, ribonucleic acid, proteins, and lipids between various cells. Some studies have reported that the components noted in exosomes in human breast milk could be transferred to the infant and cause epigenetic changes. Thus, it can affect gene expression and cellular event regulation in several tissues. Conclusions: In this manner, exosomes are associated with several pathways, including the immune system, oxidative stress, and cell cycle, and they can affect the short- and long-term health of infants. However, there is still much to learn about the functions, effectiveness, and certain impacts on the health of human breast milk exosomes.
Collapse
Affiliation(s)
- Elif Çelik
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Süleyman Demirel University, Isparta 32260, Türkiye;
| | - Özge Cemali
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Trakya University, Edirne 22030, Türkiye;
| | - Teslime Özge Şahin
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Afyonkarahisar Health Sciences University, Afyonkarahisar 03030, Türkiye;
| | - Gülsüm Deveci
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Çankırı Karatekin University, Çankırı 18100, Türkiye;
| | - Nihan Çakır Biçer
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Acıbadem Mehmet Ali Aydınlar University, Istanbul 34752, Türkiye;
| | | | - Duygu Ağagündüz
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, Ankara 06490, Türkiye
| | - Ferenc Budán
- Institute of Physiology, Medical School, University of Pécs, H-7624 Pécs, Hungary
| |
Collapse
|
9
|
Perruzza L, Heckmann J, Rezzonico Jost T, Raneri M, Guglielmetti S, Gargari G, Palatella M, Willers M, Fehlhaber B, Werlein C, Vogl T, Roth J, Grassi F, Viemann D. Postnatal supplementation with alarmins S100a8/a9 ameliorates malnutrition-induced neonate enteropathy in mice. Nat Commun 2024; 15:8623. [PMID: 39366940 PMCID: PMC11452687 DOI: 10.1038/s41467-024-52829-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 09/19/2024] [Indexed: 10/06/2024] Open
Abstract
Malnutrition is linked to 45% of global childhood mortality, however, the impact of maternal malnutrition on the child's health remains elusive. Previous studies suggested that maternal malnutrition does not affect breast milk composition. Yet, malnourished children often develop a so-called environmental enteropathy, assumed to be triggered by frequent pathogen uptake and unfavorable gut colonization. Here, we show in a murine model that maternal malnutrition induces a persistent inflammatory gut dysfunction in the offspring that establishes during nursing and does not recover after weaning onto standard diet. Early intestinal influx of neutrophils, impaired postnatal development of gut-regulatory functions, and expansion of Enterobacteriaceae were hallmarks of this enteropathy. This gut phenotype resembled those developing under deficient S100a8/a9-supply via breast milk, which is a known key factor for the postnatal development of gut homeostasis. We could confirm that S100a8/a9 is lacking in the breast milk of malnourished mothers and the offspring's intestine. Nutritional supply of S100a8 to neonates of malnourished mothers abrogated the aberrant development of gut mucosal immunity and microbiota colonization and protected them lifelong against severe enteric infections and non-infectious bowel diseases. S100a8 supplementation after birth might be a promising measure to counteract deleterious imprinting of gut immunity by maternal malnutrition.
Collapse
Affiliation(s)
- Lisa Perruzza
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), Bellinzona, Switzerland.
- Humabs BioMed SA a Subsidiary of Vir Biotechnology Inc., Bellinzona, Switzerland.
| | - Julia Heckmann
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Tanja Rezzonico Jost
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), Bellinzona, Switzerland
| | - Matteo Raneri
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), Bellinzona, Switzerland
| | - Simone Guglielmetti
- Department of Biotechnology and Biosciences (BtBs), University of Milano-Bicocca, Milan, Italy
- Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
| | - Giorgio Gargari
- Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
| | - Martina Palatella
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), Bellinzona, Switzerland
| | - Maike Willers
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Beate Fehlhaber
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | | | - Thomas Vogl
- Institute of Immunology, University of Münster, Münster, Germany
| | - Johannes Roth
- Institute of Immunology, University of Münster, Münster, Germany
| | - Fabio Grassi
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), Bellinzona, Switzerland
| | - Dorothee Viemann
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany.
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany.
- Center for Infection Research, University Würzburg, Würzburg, Germany.
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany.
| |
Collapse
|
10
|
Chehab RF, Fehr K, Moossavi S, Subbarao P, Moraes TJ, Mandhane P, de Souza RJ, Turvey SE, Khafipour E, Azad MB, Forman MR. Prenatal vitamin C and fish oil supplement use are associated with human milk microbiota composition in the Canadian CHILD Cohort Study. J Nutr Sci 2024; 13:e53. [PMID: 39345253 PMCID: PMC11428054 DOI: 10.1017/jns.2024.58] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 07/08/2024] [Accepted: 08/27/2024] [Indexed: 10/01/2024] Open
Abstract
Maternal diet may modulate human milk microbiota, but the effects of nutritional supplements are unknown. We examined the associations of prenatal diet and supplement use with milk microbiota composition. Mothers reported prenatal diet intake and supplement use using self-administered food frequency and standardised questionnaires, respectively. The milk microbiota was profiled using 16S rRNA gene sequencing. Associations of prenatal diet quality, dietary patterns, and supplement use with milk microbiota diversity and taxonomic structure were examined using Wilcoxon signed-rank tests and multivariable models adjusting for relevant confounders. A subset of 645 mothers participating in the CHILD Cohort Study (originally known as the Canadian Healthy Infant Longitudinal Development Study) provided one milk sample between 2 and 6 months postpartum and used prenatal multivitamin supplements ≥4 times a week. After adjusting for confounders, vitamin C supplement use was positively associated with milk bacterial Shannon diversity (β = 0.18, 95% CI = 0.05, 0.31) and Veillonella and Granulicatella relative abundance (β = 0.54; 95% CI = 0.05, 1.03 and β = 0.44; 95% CI = 0.04, 0.84, respectively), and negatively associated with Finegoldia relative abundance (β = -0.31; 95% CI = -0.63, -0.01). Fish oil supplement use was positively associated with Streptococcus relative abundance (β = 0.26; 95% CI = 0.03, 0.50). Prenatal diet quality and dietary patterns were not associated with milk microbiota composition. Prenatal vitamin C and fish oil supplement use were associated with differences in the milk microbiota composition. Future studies are needed to confirm our findings and elucidate mechanisms linking maternal supplement use to milk microbiota and child health.
Collapse
Affiliation(s)
- Rana F Chehab
- Department of Nutrition Science, Purdue University, West Lafayette, IN, USA
| | - Kelsey Fehr
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
| | - Shirin Moossavi
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
- Department of Pediatrics, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, University of Calgary, Calgary, AB, Canada
| | - Padmaja Subbarao
- Hospital for Sick Children, Department of Pediatrics & Physiology and Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Theo J Moraes
- Department of Pediatrics, Hospital for Sick Children and University of Toronto, Toronto, ON, Canada
| | - Piushkumar Mandhane
- Department of Paediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Russell J de Souza
- Department of Health Research Methods, Evidence, and Impact, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
- Population Health Research Institute, Hamilton Health Sciences Corporation, Hamilton, ON, Canada
| | - Stuart E Turvey
- Department of Pediatrics, British Columbia Children's Hospital and The University of British Columbia, Vancouver, BC, Canada
| | - Ehsan Khafipour
- Department of Animal Science, University of Manitoba, Winnipeg, MB, Canada
| | - Meghan B Azad
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
- Manitoba Interdisciplinary Lactation Centre (MILC), Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Michele R Forman
- Department of Nutrition Science, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
11
|
Gerçel G, Anadolulu AI. Neonatal Gastrointestinal Perforations: A 4-year Experience in a Single Centre. Afr J Paediatr Surg 2024:01434821-990000000-00016. [PMID: 39254062 DOI: 10.4103/ajps.ajps_96_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 01/05/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND Gastrointestinal perforation (GIP) during the neonatal period is still a significant problem despite improved neonatal care. The study aimed to report on incidence, management, morbidity and mortality. MATERIAL AND METHODS Records of neonates with GIPs between October 2018 and November 2022 were retrospectively analysed. RESULTS There were 47 patients, 22 (46.8%) males and 25 (53.2%) females. The incidence of neonatal GIP was 0.39% amongst all newborns treated in the neonatal intensive care unit. The mean gestational age was 30.4 ± 4.5 (23-38) weeks, and the mean birth weight was 1493.08 ± 753 (580-2940) g. Of 47 neonates, 5 (10.6%) were full term and 42 (89.4%) were preterm. The mean age of surgery was 12.25 ± 9.89 (0-41) days. A laparotomy was performed in 43 (91.4%) of 47 neonates, while seven of the patients underwent surgical intervention after decompression by percutaneous drainage. Four patients were managed with peritoneal drainage alone due to poor general condition. The pathologies unrelated to necrotising enterocolitis (NEC) were the most common cause of GIPs (55.3%) and included spontaneous intestinal perforation (n = 18), stomach perforation (n = 4), segmental volvulus (n = 2), acute mesenteric ischaemia (n = 1) and meconium peritonitis (n = 1). Overall survival was 55.4%. CONCLUSION GIPs are one of the most significant causes of mortality in newborns. The most common cause of perforations is non-NEC entities and can be seen in the entire intestinal system from the stomach to the colon. Surgical exploration is still the primary management model.
Collapse
Affiliation(s)
- Gonca Gerçel
- Department of Pediatric Surgery, Şanlıurfa Training and Research Hospital, Şanlıurfa, Turkey
- Department of Pediatric Surgery, İstanbul Sancaktepe Şehit Prof. Dr. İlhan Varank Training and Research Hospital, Istanbul, Turkey
| | - Ali Ihsan Anadolulu
- Department of Pediatric Surgery, Mehmet Akif İnan Training and Research Hospital, Şanlıurfa, Turkey
- Department of Pediatric Surgery, Faculty of Medicine, Goztepe Prof. Dr. Suleyman Yalcin City Hospital, Istanbul Medeniyet University, Istanbul, Turkey
| |
Collapse
|
12
|
Vassilopoulou E, Agostoni C, Feketea G, Alberti I, Gianni ML, Milani GP. The Role of Breastfeeding in Acute Respiratory Infections in Infancy. Pediatr Infect Dis J 2024:00006454-990000000-00942. [PMID: 38986006 DOI: 10.1097/inf.0000000000004454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
BACKGROUND Acute respiratory infections (ARIs) affect the respiratory tract, are often caused by viruses such as respiratory syncytial virus and rhinovirus, and present symptoms such as coughing, fever, respiratory distress, and breathing difficulty. The global adherence to exclusive breastfeeding (BF) for the first 6 months of life has reached 44%, supported by the World Health Organization and United Nations International Children's Emergency Fund efforts. BF provides vital nutrients and contributes to infant immune system development, protecting against infections. The role of BF in preventing and reducing complications of ARIs in infants is gaining attention, prompting a review of current data and future research needs. This review aims to summarize the evidence on the role of BF in reducing the risk and severity of ARIs in infants, elucidate the adaptations in breast milk composition during infections, and identify relevant research needs. METHODS AND RESULTS Human milk (HM) is rich in immunoglobulins, antimicrobial peptides, and immunomodulatory factors that protect against various pathogens, including respiratory viruses. Several studies have demonstrated that BF is associated with a significant reduction in hospitalization, oxygen requirements, and mortality in infants with ARIs. The effectiveness of BF varies according to the specific respiratory virus, and a longer duration of exclusive BF appears to enhance its protective effect. It is documented that the composition of HM adjusts dynamically in response to infections, fortifying the infant's immune defenses. Specific immunological components of HM, including leukocytes and immunoglobulins, increase in response to infection in the infant, contributing to the enhancement of the immune defense in infants. Immune-boosting microRNAs enhance immune transfer to the infants and promote early gut maturation, and the HM microbiome along with other factors modifies the infant's gut microbiome and immune system. CONCLUSIONS BF defends infants from respiratory infections, and the investigation of the microRNAs in HM offers new insights into its antiviral properties. The promotion of BF, especially in vulnerable communities, is of paramount importance in alleviating the global burden of ARIs in infancy.
Collapse
Affiliation(s)
- Emilia Vassilopoulou
- From the Pediatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Nutritional Sciences and Dietetics, International Hellenic University, Thessaloniki, Greece
| | - Carlo Agostoni
- From the Pediatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Gavriela Feketea
- Department of Pharmacology, Toxicology and Clinical Pharmacology, University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Pediatric Allergy Outpatient Clinic, Department of Pediatrics, "Karamandaneio" Children's Hospital of Patra, Patras, Greece
| | - Ilaria Alberti
- From the Pediatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Maria Lorella Gianni
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
- Fondazione I.R.C.C.S. Ca' Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit, Milan, Italy
| | - Gregorio Paolo Milani
- From the Pediatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
13
|
Lin F, Luo J, Zhu Y, Liang H, Li D, Han D, Chang Q, Pan P, Zhang Y. Association Between Adverse Early Life Factors and Telomere Length in Middle and Late Life. Innov Aging 2024; 8:igae070. [PMID: 39350941 PMCID: PMC11441326 DOI: 10.1093/geroni/igae070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Indexed: 10/04/2024] Open
Abstract
Background and Objectives Telomere length (TL) has been acknowledged as biomarker of biological aging. Numerous investigations have examined associations between individual early life factors and leukocyte TL; however, the findings were far from consistent. Research Design and Methods We evaluated the relationship between individual and combined early life factors and leukocytes TL in middle and late life using data from the UK Biobank. The early life factors (eg, maternal smoking, breastfeeding, birth weight, and comparative body size and height to peers at age 10) were measured. The regression coefficients (β) and 95% confidence interval (CI) were applied to assess the link of the early life factors and TL in adulthood. Flexible parametric survival models incorporated age to calculate the relationship between early life factors and life expectancy. Results Exposure to maternal smoking, lack of breastfeeding, low birth weight, and shorter height compared to peers at age 10 were identified to be associated with shorter TL in middle and older age according to the large population-based study with 197 504 participants. Individuals who experienced more than 3 adverse early life factors had the shortest TL in middle and late life (β = -0.053; 95% CI = -0.069 to -0.038; p < .0001), as well as an average of 0.54 years of life loss at the age of 45 and 0.49 years of life loss at the age of 60, compared to those who were not exposed to any early life risk factors. Discussion and Implications Early life factors including maternal smoking, non-breastfed, low birth weight, and shorter height compared to peers at age 10 were associated with shorter TL in later life. In addition, an increased number of the aforementioned factors was associated with a greater likelihood of shorter TL in adulthood, as well as a reduced life expectancy.
Collapse
Affiliation(s)
- Fengyu Lin
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiefeng Luo
- Department of Gynecology and Obstetrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- International Collaborative Research Center for Medical Metabolomics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yiqun Zhu
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China
| | - Huaying Liang
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Dianwu Li
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Duoduo Han
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qinyu Chang
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Pinhua Pan
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- FuRong Laboratory, Central South University, Changsha, Hunan, China
| | - Yan Zhang
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
14
|
Gu H, Tao E, Fan Y, Long G, Jia X, Yuan T, Chen L, Shu X, Zheng W, Jiang M. Effect of β-lactam antibiotics on the gut microbiota of term neonates. Ann Clin Microbiol Antimicrob 2024; 23:69. [PMID: 39113137 PMCID: PMC11308410 DOI: 10.1186/s12941-024-00730-2] [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: 04/27/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
β-Lactam antibiotics are a class of antibiotics commonly used to treat bacterial infections. However, the effects of β-lactam antibiotics on term neonatal intestinal flora have not been fully elucidated. Hospitalized full-term newborns receiving β-lactam antibiotics formed the antibiotic group (n = 67), while those without antibiotic treatment comprised the non-antibiotic group (n = 47). A healthy group included healthy full-term newborns (n = 16). Stool samples were collected for 16 S rDNA sequencing to analyze gut microbiota variations. Further investigation was carried out within the β-lactam antibiotic group, exploring the effects of antibiotic use on the newborns' gut microbiota in relation to the duration and type of antibiotic administration, delivery method, and feeding practices. The antibiotic group exhibited significant difference of microbial community composition compared to the other groups. Genera like Klebsiella, Enterococcus, Streptococcus, Alistipes, and Aeromonas were enriched, while Escherichia-Shigella, Clostridium sensu stricto 1, Bifidobacterium, and Parabacteroides were reduced. Klebsiella negatively correlated with Escherichia-Shigella, positively with Enterobacter, while Escherichia-Shigella negatively correlated with Enterococcus and Streptococcus. Regardless of neonatal age, β-lactam antibiotics induced an elevated abundance of Klebsiella and Enterococcus. The impact on gut microbiota varied with the duration and type of antibiotic (cefotaxime or ampicillin/sulbactam). Compared to vaginal delivery, cesarean delivery after β-lactam treatment heightened the abundance of Klebsiella, Enterobacteriaceae_Unclassified, Lactobacillales_Unclassified, and Pectobacterium. Feeding patterns minimally influenced β-lactam-induced alterations. In conclusion, β-lactam antibiotic treatment for neonatal pneumonia and sepsis markedly disrupted intestinal microbiota, favoring Klebsiella, Enterococcus, Streptococcus, Alistipes, and Aeromonas. The impact of β-lactam varied by duration, type, and delivery method, emphasizing heightened disruptions post-cesarean delivery.
Collapse
Affiliation(s)
- Hongdan Gu
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
- Department of Pediatrics, Wenling Maternal and Child Health Care Hospital, Wenling, Zhejiang, 317500, China
| | - Enfu Tao
- Department of Neonatology and NICU, Wenling Maternal and Child Health Care Hospital, Wenling, Zhejiang, 317500, China
| | - Yijia Fan
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Gao Long
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Xinyi Jia
- Department of Gastroenterology and Pediatric Endoscopy Center, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Children's Hospital, Zhejiang University School of Medicine, No. 3333, Binsheng Road, Hangzhou, Zhejiang, 310052, China
| | - Tianming Yuan
- Department of Neonatology, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310052, China
| | - Lihua Chen
- Department of Neonatology, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310052, China
| | - Xiaoli Shu
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Wei Zheng
- Department of Gastroenterology and Pediatric Endoscopy Center, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Children's Hospital, Zhejiang University School of Medicine, No. 3333, Binsheng Road, Hangzhou, Zhejiang, 310052, China
| | - Mizu Jiang
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China.
- Department of Gastroenterology and Pediatric Endoscopy Center, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Children's Hospital, Zhejiang University School of Medicine, No. 3333, Binsheng Road, Hangzhou, Zhejiang, 310052, China.
| |
Collapse
|
15
|
Cao X, Fang Y, Bandan P, Suo L, Jiacuo G, Wu Y, Cuoji A, Zhuoga D, Chen Y, Ji D, Quzhen C, Zhang K. Age-specific composition of milk microbiota in Tibetan sheep and goats. Appl Microbiol Biotechnol 2024; 108:411. [PMID: 38980443 PMCID: PMC11233330 DOI: 10.1007/s00253-024-13252-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 07/02/2024] [Accepted: 07/03/2024] [Indexed: 07/10/2024]
Abstract
This study investigates the dynamic changes in milk nutritional composition and microbial communities in Tibetan sheep and goats during the first 56 days of lactation. Milk samples were systematically collected at five time points (D0, D7, D14, D28, D56) post-delivery. In Tibetan sheep, milk fat, protein, and casein contents were highest on D0, gradually decreased, and stabilized after D14, while lactose and galactose levels showed the opposite trend. Goat milk exhibited similar initial peaks, with significant changes particularly between D0, D7, D14, and D56. 16S rRNA gene sequencing revealed increasing microbial diversity in both species over the lactation period. Principal coordinates analysis identified distinct microbial clusters corresponding to early (D0-D7), transitional (D14-D28), and mature (D56) stages. Core phyla, including Proteobacteria, Firmicutes, Bacteroidetes, and Actinobacteria, dominated the milk microbiota, with significant temporal shifts. Core microbes like Lactobacillus, Leuconostoc, and Streptococcus were common in both species, with species-specific taxa observed (e.g., Pediococcus in sheep, Shewanella in goats). Furthermore, we observed a highly shared core microbiota in sheep and goat milk, including Lactobacillus, Leuconostoc, and Streptococcus. Spearman correlation analysis highlighted significant relationships between specific microbial genera and milk nutrients. For instance, Lactobacillus positively correlated with total solids, non-fat milk solids, protein, and casein, while Mannheimia negatively correlated with protein content. This study underscores the complex interplay between milk composition and microbial dynamics in Tibetan sheep and goats, informing strategies for livestock management and nutritional enhancement. KEY POINTS: • The milk can be classified into three types based on the microbiota composition • The changes of milk microbiota are closely related to the variations in nutrition • Filter out microbiota with species specificity and age specificity in the milk.
Collapse
Affiliation(s)
- Xi Cao
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, 712100, Yangling, China
| | - Yumeng Fang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, 712100, Yangling, China
| | - Pingcuo Bandan
- Institute of Animal Sciences, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, 850009, China
| | - Langda Suo
- Institute of Animal Sciences, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, 850009, China
| | - Gesang Jiacuo
- Institute of Animal Sciences, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, 850009, China
| | - Yujiang Wu
- Institute of Animal Sciences, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, 850009, China
| | - Awang Cuoji
- Institute of Animal Sciences, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, 850009, China
| | - Deqing Zhuoga
- Institute of Animal Sciences, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, 850009, China
| | - Yulin Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, 712100, Yangling, China
| | - De Ji
- Institute of Animal Sciences, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, 850009, China.
| | - Ciren Quzhen
- Institute of Animal Sciences, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, 850009, China.
| | - Ke Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, 712100, Yangling, China.
| |
Collapse
|
16
|
Moon S, Lee KW, Park M, Moon J, Park SH, Kim S, Hwang J, Yoon JW, Jeon SM, Kim JS, Jeon YJ, Kweon DH. 3-Fucosyllactose-mediated modulation of immune response against virus infection. Int J Antimicrob Agents 2024; 64:107187. [PMID: 38697577 DOI: 10.1016/j.ijantimicag.2024.107187] [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: 11/06/2023] [Revised: 03/19/2024] [Accepted: 04/24/2024] [Indexed: 05/05/2024]
Abstract
Viral pathogens, particularly influenza and SARS-CoV-2, pose a significant global health challenge. Given the immunomodulatory properties of human milk oligosaccharides, in particular 2'-fucosyllactose and 3-fucosyllactose (3-FL), we investigated their dietary supplementation effects on antiviral responses in mouse models. This study revealed distinct immune modulations induced by 3-FL. RNA-sequencing data showed that 3-FL increased the expression of interferon receptors, such as Interferon Alpha and Beta Receptor (IFNAR) and Interferon Gamma Receptor (IFNGR), while simultaneously downregulating interferons and interferon-stimulated genes, an effect not observed with 2'-fucosyllactose supplementation. Such modulation enhanced antiviral responses in both cell culture and animal models while attenuating pre-emptive inflammatory responses. Nitric oxide concentrations in 3-FL-supplemented A549 cells and mouse lung tissues were elevated exclusively upon infection, reaching 5.8- and 1.9-fold increases over control groups, respectively. In addition, 3-FL promoted leukocyte infiltration into the site of infection upon viral challenge. 3-FL supplementation provided protective efficacy against lethal influenza challenge in mice. The demonstrated antiviral efficacy spanned multiple influenza strains and extended to SARS-CoV-2. In conclusion, 3-FL is a unique immunomodulator that helps protect the host from viral infection while suppressing inflammation prior to infection.
Collapse
Affiliation(s)
- Seokoh Moon
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Ki Wook Lee
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Myungseo Park
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jeonghui Moon
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Sang Hee Park
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Soomin Kim
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jaehyeon Hwang
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jong-Won Yoon
- Advanced Protein Technologies Corp., Suwon, Republic of Korea
| | - Seon-Min Jeon
- Advanced Protein Technologies Corp., Suwon, Republic of Korea
| | - Jun-Seob Kim
- Department of Nano-Bioengineering, Incheon National University, Incheon, Republic of Korea.
| | - Young-Jun Jeon
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea.
| | - Dae-Hyuk Kweon
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea; Advanced Protein Technologies Corp., Suwon, Republic of Korea.
| |
Collapse
|
17
|
Martínez-Martínez AB, Lamban-Per BM, Lezaun M, Rezusta A, Arbones-Mainar JM. Exploring Functional Products and Early-Life Dynamics of Gut Microbiota. Nutrients 2024; 16:1823. [PMID: 38931178 PMCID: PMC11206896 DOI: 10.3390/nu16121823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 05/31/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Research on the microbiome has progressed from identifying specific microbial communities to exploring how these organisms produce and modify metabolites that impact a wide range of health conditions, including gastrointestinal, metabolic, autoimmune, and neurodegenerative diseases. This review provides an overview of the bacteria commonly found in the intestinal tract, focusing on their main functional outputs. We explore biomarkers that not only indicate a well-balanced microbiota but also potential dysbiosis, which could foreshadow susceptibility to future health conditions. Additionally, it discusses the establishment of the microbiota during the early years of life, examining factors such as gestational age at birth, type of delivery, antibiotic intake, and genetic and environmental influences. Through a comprehensive analysis of current research, this article aims to enhance our understanding of the microbiota's foundational development and its long-term implications for health and disease management.
Collapse
Affiliation(s)
- Ana B. Martínez-Martínez
- Facultad de Ciencias de la Salud, Universidad de Zaragoza, 50009 Zaragoza, Spain;
- Instituto de Investigación Sanitaria Aragón, 50009 Zaragoza, Spain;
| | - Belen M. Lamban-Per
- Department of Clinical Microbiology, Miguel Servet University Hospital, 50009 Zaragoza, Spain; (B.M.L.-P.); (M.L.)
| | - Maria Lezaun
- Department of Clinical Microbiology, Miguel Servet University Hospital, 50009 Zaragoza, Spain; (B.M.L.-P.); (M.L.)
| | - Antonio Rezusta
- Instituto de Investigación Sanitaria Aragón, 50009 Zaragoza, Spain;
- Department of Clinical Microbiology, Miguel Servet University Hospital, 50009 Zaragoza, Spain; (B.M.L.-P.); (M.L.)
| | - Jose M. Arbones-Mainar
- Department of Clinical Microbiology, Miguel Servet University Hospital, 50009 Zaragoza, Spain; (B.M.L.-P.); (M.L.)
- Adipocyte and Fat Biology Laboratory (AdipoFat), Instituto Aragonés de Ciencias de la Salud (IACS), 50009 Zaragoza, Spain
- CIBER Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
18
|
Freitas RGBON, Vasques ACJ, da Rocha Fernandes G, Ribeiro FB, Solar I, Shivappa N, Hébert JR, de Almeida-Pititto B, Geloneze B, Ferreira SRG. Gut bacterial markers involved in association of dietary inflammatory index with visceral adiposity. Nutrition 2024; 122:112371. [PMID: 38430843 DOI: 10.1016/j.nut.2024.112371] [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: 08/06/2023] [Revised: 11/14/2023] [Accepted: 01/24/2024] [Indexed: 03/05/2024]
Abstract
OBJECTIVE To deepen the understanding of the influence of diet on weight gain and metabolic disturbances, we examined associations between diet-related inflammation and body composition and fecal bacteria abundances in participants of the Nutritionists' Health Study. METHODS Early-life, dietary and clinical data were obtained from 114 women aged ≤45 years. A validated food frequency questionnaire was used to calculate the energy-adjusted dietary inflammatory index (E-DII). Participants' data were compared by E-DII quartiles using ANOVA or Kruskal-Wallis. Associations of DXA-determined body composition with the E-DII were tested by multiple linear regression using DAG-oriented adjustments. Fecal microbiota was analyzed targeting the V4 region of the 16S rRNA gene. Spearman correlation coefficients were used to test linear associations; differential abundance of genera across the E-DII quartiles was assessed by pair-wise comparisons. RESULTS E-DII score was associated with total fat (b=1.80, p<0.001), FMI (b=0.08, p<0.001) and visceral fat (b=1.19, p=0.02), independently of maternal BMI, birth type and breastfeeding. E-DII score was directly correlated to HOMA-IR (r=0.30; p=0.004), C-reactive protein (r=0.29; p=0.003) and to the abundance of Actinomyces, and inversely correlated to the abundance of Eubacterium.xylanophilum.group. Actinomyces were significantly more abundant in the highest (most proinflammatory) E-DII quartile. CONCLUSIONS Association of E-DII with markers of insulin resistance, inflammation, body adiposity and certain gut bacteria are consistent with beneficial effects of anti-inflammatory diet on body composition and metabolic profile. Bacterial markers, such as Actinomyces, could be involved in the association between the dietary inflammation with visceral adiposity. Studies designed to explore how a pro-inflammatory diet affects both central fat deposition and gut microbiota are needed.
Collapse
Affiliation(s)
- Renata G B O N Freitas
- Department of Epidemiology, School of Public Health, University of São Paulo, Brazil; Laboratory of Investigation in Metabolism and Diabetes, School of Medical Sciences, University of Campinas, Brazil
| | - Ana Carolina J Vasques
- Laboratory of Investigation in Metabolism and Diabetes, School of Medical Sciences, University of Campinas, Brazil; School of Applied Sciences, University of Campinas, São Paulo, Brazil
| | | | - Francieli B Ribeiro
- Laboratory of Investigation in Metabolism and Diabetes, School of Medical Sciences, University of Campinas, Brazil; School of Applied Sciences, University of Campinas, São Paulo, Brazil
| | - Isabela Solar
- Laboratory of Investigation in Metabolism and Diabetes, School of Medical Sciences, University of Campinas, Brazil; School of Applied Sciences, University of Campinas, São Paulo, Brazil
| | - Nitin Shivappa
- Cancer Prevention and Control Program, Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC; Department of Nutrition, Connecting Health Innovations, Columbia, SC
| | - James R Hébert
- Cancer Prevention and Control Program, Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC; Department of Nutrition, Connecting Health Innovations, Columbia, SC
| | | | - Bruno Geloneze
- Laboratory of Investigation in Metabolism and Diabetes, School of Medical Sciences, University of Campinas, Brazil; Obesity and Comorbidities Research Center, University of Campinas, São Paulo, Brazil
| | | |
Collapse
|
19
|
Zhang S, Wang Q, Tan DEL, Sikka V, Ng CH, Xian Y, Li D, Muthiah M, Chew NWS, Storm G, Tong L, Wang J. Gut-liver axis: Potential mechanisms of action of food-derived extracellular vesicles. J Extracell Vesicles 2024; 13:e12466. [PMID: 38887165 PMCID: PMC11183959 DOI: 10.1002/jev2.12466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 05/03/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024] Open
Abstract
Food-derived extracellular vesicles (FEVs) are nanoscale membrane vesicles obtained from dietary materials such as breast milk, plants and probiotics. Distinct from other EVs, FEVs can survive the harsh degrading conditions in the gastrointestinal tract and reach the intestines. This unique feature allows FEVs to be promising prebiotics in health and oral nanomedicine for gut disorders, such as inflammatory bowel disease. Interestingly, therapeutic effects of FEVs have recently also been observed in non-gastrointestinal diseases. However, the mechanisms remain unclear or even mysterious. It is speculated that orally administered FEVs could enter the bloodstream, reach remote organs, and thus exert therapeutic effects therein. However, emerging evidence suggests that the amount of FEVs reaching organs beyond the gastrointestinal tract is marginal and may be insufficient to account for the significant therapeutic effects achieved regarding diseases involving remote organs such as the liver. Thus, we herein propose that FEVs primarily act locally in the intestine by modulating intestinal microenvironments such as barrier integrity and microbiota, thereby eliciting therapeutic impact remotely on the liver in non-gastrointestinal diseases via the gut-liver axis. Likewise, drugs delivered to the gastrointestinal system through FEVs may act via the gut-liver axis. As the liver is the main metabolic hub, the intestinal microenvironment may be implicated in other metabolic diseases. In fact, many patients with non-alcoholic fatty liver disease, obesity, diabetes and cardiovascular disease suffer from a leaky gut and dysbiosis. In this review, we provide an overview of the recent progress in FEVs and discuss their biomedical applications as therapeutic agents and drug delivery systems, highlighting the pivotal role of the gut-liver axis in the mechanisms of action of FEVs for the treatment of gut disorders and metabolic diseases.
Collapse
Affiliation(s)
- Sitong Zhang
- Department of Surgery, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Qiyue Wang
- Jinan Central HospitalShandong First Medical University & Shandong Academy of Medical SciencesJinanChina
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanChina
| | - Daniel En Liang Tan
- Department of Surgery, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Vritika Sikka
- Department of Surgery, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Cheng Han Ng
- Division of Gastroenterology and Hepatology, Department of MedicineNational University HospitalSingaporeSingapore
| | - Yan Xian
- Department of Surgery, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Dan Li
- Department of Food Science and Technology, Faculty of ScienceNational University of SingaporeSingaporeSingapore
| | - Mark Muthiah
- Division of Gastroenterology and Hepatology, Department of MedicineNational University HospitalSingaporeSingapore
- National University Centre for Organ TransplantationNational University Health SystemSingaporeSingapore
| | - Nicholas W. S. Chew
- Department of CardiologyNational University Heart CentreNational University Health SystemSingaporeSingapore
| | - Gert Storm
- Department of Surgery, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Lingjun Tong
- Jinan Central HospitalShandong First Medical University & Shandong Academy of Medical SciencesJinanChina
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanChina
| | - Jiong‐Wei Wang
- Department of Surgery, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Cardiovascular Research Institute (CVRI)National University Heart Centre Singapore (NUHCS)SingaporeSingapore
- Department of Physiology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| |
Collapse
|
20
|
Zhang M, Qiao H, Yang S, Kwok LY, Zhang H, Zhang W. Human Breast Milk: The Role of Its Microbiota and Metabolites in Infant Health. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:10665-10678. [PMID: 38691667 DOI: 10.1021/acs.jafc.3c07690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
This review explores the role of microorganisms and metabolites in human breast milk and their impact on neonatal health. Breast milk serves as both a primary source of nutrition for newborns and contributes to the development and maturation of the digestive, immunological, and neurological systems. It has the potential to reduce the risks of infections, allergies, and asthma. As our understanding of the properties of human milk advances, there is growing interest in incorporating its benefits into personalized infant nutrition strategies, particularly in situations in which breastfeeding is not an option. Future infant formula products are expected to emulate the composition and advantages of human milk, aligning with an evolving understanding of infant nutrition. The long-term health implications of human milk are still under investigation.
Collapse
Affiliation(s)
- Meng Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Hui Qiao
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Shuwei Yang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Lai-Yu Kwok
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Heping Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Wenyi Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| |
Collapse
|
21
|
Adeniyi-Ipadeola GO, Hoffman KL, Yang H, Javornik Cregeen SJ, Preidis GA, Ramani S, Hair AB. Human milk cream alters intestinal microbiome of preterm infants: a prospective cohort study. Pediatr Res 2024; 95:1564-1571. [PMID: 38228744 DOI: 10.1038/s41390-023-02948-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 11/09/2023] [Accepted: 11/18/2023] [Indexed: 01/18/2024]
Abstract
BACKGROUND In very low birth weight (VLBW) infants, human milk cream added to standard human milk fortification is used to improve growth. This study aimed to evaluate the impact of cream supplement on the intestinal microbiome of VLBW infants. METHODS Whole genome shotgun sequencing was performed on stool (n = 57) collected from a cohort of 23 infants weighing 500-1250 grams (control = 12, cream = 11). Both groups received an exclusive human milk diet (mother's own milk, donor human milk, and donor human milk-derived fortifier) with the cream group receiving an additional 2 kcal/oz cream at 100 mL/kg/day of fortified feeds and then 4 kcal/oz if poor growth. RESULTS While there were no significant differences in alpha diversity, infants receiving cream significantly differed from infants in the control group in beta diversity. Cream group samples had significantly higher prevalence of Proteobacteria and significantly lower Firmicutes compared to control group. Klebsiella species dominated the microbiota of cream-exposed infants, along with bacterial pathways involved in lipid metabolism and metabolism of cofactors and amino acids. CONCLUSIONS Cream supplementation significantly altered composition of the intestinal microbiome of VLBW infants to favor increased prevalence of Proteobacteria and functional gene content associated with these bacteria. IMPACT We report changes to the intestinal microbiome associated with administration of human milk cream; a novel supplement used to improve growth rates of preterm very low birth weight infants. Since little is known about the impact of cream on intestinal microbiota composition of very low birth weight infants, our study provides valuable insight on the effects of diet on the microbiome of this population. Dietary supplements administered to preterm infants in neonatal intensive care units have the potential to influence the intestinal microbiome composition which may affect overall health status of the infant.
Collapse
Affiliation(s)
- Grace O Adeniyi-Ipadeola
- Graduate Program in Immunology & Microbiology, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Kristi L Hoffman
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Heeju Yang
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | - Sara J Javornik Cregeen
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Geoffrey A Preidis
- Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| | - Sasirekha Ramani
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Amy B Hair
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA.
| |
Collapse
|
22
|
Shahi S, Kang T, Fonseka P. Extracellular Vesicles in Pathophysiology: A Prudent Target That Requires Careful Consideration. Cells 2024; 13:754. [PMID: 38727289 PMCID: PMC11083420 DOI: 10.3390/cells13090754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Extracellular vesicles (EVs) are membrane-bound particles released by cells to perform multitudes of biological functions. Owing to their significant implications in diseases, the pathophysiological role of EVs continues to be extensively studied, leading research to neglect the need to explore their role in normal physiology. Despite this, many identified physiological functions of EVs, including, but not limited to, tissue repair, early development and aging, are attributed to their modulatory role in various signaling pathways via intercellular communication. EVs are widely perceived as a potential therapeutic strategy for better prognosis, primarily through utilization as a mode of delivery vehicle. Moreover, disease-associated EVs serve as candidates for the targeted inhibition by pharmacological or genetic means. However, these attempts are often accompanied by major challenges, such as off-target effects, which may result in adverse phenotypes. This renders the clinical efficacy of EVs elusive, indicating that further understanding of the specific role of EVs in physiology may enhance their utility. This review highlights the essential role of EVs in maintaining cellular homeostasis under different physiological settings, and also discusses the various aspects that may potentially hinder the robust utility of EV-based therapeutics.
Collapse
Affiliation(s)
| | | | - Pamali Fonseka
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia; (S.S.); (T.K.)
| |
Collapse
|
23
|
Ding M, Li B, Chen H, Ross RP, Stanton C, Zhao J, Chen W, Yang B. Bifidobacterium longum Subsp. infantis Promotes IgA Level of Growing Mice in a Strain-Specific and Intestinal Niche-Dependent Manner. Nutrients 2024; 16:1148. [PMID: 38674840 PMCID: PMC11054607 DOI: 10.3390/nu16081148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/06/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Throughout infancy, IgA is crucial for maintaining gut mucosal immunity. This study aims to determine whether supplementing newborn mice with eight different strains of Bifidobacterium longum subsp. infantis might regulate their IgA levels. The strains were gavaged to BALB/C female (n = 8) and male (n = 8) dams at 1-3 weeks old. Eight strains of B. longum subsp. infantis had strain-specific effects in the regulation of intestinal mucosal barriers. B6MNI, I4MI, and I10TI can increase the colonic IgA level in females and males. I8TI can increase the colonic IgA level in males. B6MNI was also able to significantly increase the colonic sIgA level in females. B6MNI, I4MI, I8TI, and I10TI regulated colonic and Peyer's patch IgA synthesis genes but had no significant effect on IgA synthesis pathway genes in the jejunum and ileum. Moreover, the variety of sIgA-coated bacteria in male mice was changed by I4MI, I5TI, I8TI, and B6MNI. These strains also can decrease the relative abundance of Escherichia coli. These results indicate that B. longum subsp. infantis can promote IgA levels but show strain specificity. Different dietary habits with different strains of Bifidobacterium may have varying effects on IgA levels when supplemented in early infancy.
Collapse
Affiliation(s)
- Mengfan Ding
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (M.D.); (B.L.); (H.C.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Bowen Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (M.D.); (B.L.); (H.C.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Haiqin Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (M.D.); (B.L.); (H.C.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Reynolds Paul Ross
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi 214122, China; (R.P.R.); (C.S.)
- APC Microbiome Ireland, University College Cork, T12 R229 Cork, Ireland
| | - Catherine Stanton
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi 214122, China; (R.P.R.); (C.S.)
- APC Microbiome Ireland, University College Cork, T12 R229 Cork, Ireland
- Teagasc Food Research Centre, Moorepark, Fermoy, P61 C996 Cork, Ireland
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (M.D.); (B.L.); (H.C.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (M.D.); (B.L.); (H.C.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Bo Yang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (M.D.); (B.L.); (H.C.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi 214122, China; (R.P.R.); (C.S.)
| |
Collapse
|
24
|
Alba C, Carrera M, Álvarez-Calatayud G, Arroyo R, Fernández L, Rodríguez JM. Evaluation of Safety and Beneficial Health Effects of the Human-Milk Strain Bifidobacterium breve DSM32583: An Infant Pilot Trial. Nutrients 2024; 16:1134. [PMID: 38674825 PMCID: PMC11053739 DOI: 10.3390/nu16081134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/05/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Human milk promotes the growth of bifidobacteria in the infant gut. Adding bifidobacterial species to infant formula may contribute to increasing their presence in the gut of formula-fed infants. Therefore, the safety and anti-infectious effects of Bifidobacterium breve DSM32583, a breast milk isolate, were assessed in a pilot trial involving 3-month-old infants. The infants were randomly assigned to either the probiotic (PG) or the control (CG) groups. All the infants consumed the same formula, although it was supplemented with the strain (1 × 107 cfu/g of formula) in the PG. Overall, 160 infants (80 per group) finished the intervention. Infants in CG gained more weight compared to PG (p < 0.05), but the weights for age Z-scores at 6 months were within the normal distribution for this age group. The rates of infections affecting the gastrointestinal and respiratory tracts and antibiotic therapy were significantly lower in the PG. The bifidobacterial population and the level of short-chain fatty acids were higher (p < 0.05) in the fecal samples of PG infants. No adverse events related to formula consumption were observed. In conclusion, the administration of an infant formula with B. breve DSM32583 was safe and exerted potential beneficial effects on gut health.
Collapse
Affiliation(s)
- Claudio Alba
- Department of Nutrition and Food Science, Complutense University of Madrid, 28040 Madrid, Spain; (C.A.); (R.A.)
| | - Marta Carrera
- Centro de Atención Primaria Silvano, Comunidad de Madrid, 28043 Madrid, Spain;
| | | | - Rebeca Arroyo
- Department of Nutrition and Food Science, Complutense University of Madrid, 28040 Madrid, Spain; (C.A.); (R.A.)
| | - Leónides Fernández
- Department of Galenic Pharmacy and Food Technology, Complutense University of Madrid, 28040 Madrid, Spain;
| | - Juan M. Rodríguez
- Department of Nutrition and Food Science, Complutense University of Madrid, 28040 Madrid, Spain; (C.A.); (R.A.)
| |
Collapse
|
25
|
Wiley KS, Gregg AM, Fox MM, Lagishetty V, Sandman CA, Jacobs JP, Glynn LM. Contact with caregivers is associated with composition of the infant gastrointestinal microbiome in the first 6 months of life. AMERICAN JOURNAL OF BIOLOGICAL ANTHROPOLOGY 2024; 183:e24858. [PMID: 37804008 PMCID: PMC10922139 DOI: 10.1002/ajpa.24858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/18/2023] [Accepted: 09/24/2023] [Indexed: 10/08/2023]
Abstract
OBJECTIVES Little is known about how physical contact at birth and early caregiving environments influence the colonization of the infant gastrointestinal microbiome. We investigated how infant contact with caregivers at birth and within the first 2 weeks of life relates to the composition of the gastrointestinal microbiome in a sample of U.S. infants (n = 60). METHODS Skin-to-skin and physical contact with caregivers at birth and early caregiving environments were surveyed at 2 weeks postpartum. Stool samples were collected from infants at 2 weeks, 2, 6, and 12 months of age and underwent 16S rRNA sequencing as a proxy for the gastrointestinal microbiome. Associations between early caregiving environments and alpha and beta diversity, and differential abundance of bacteria at the genus level were assessed using PERMANOVA, and negative binomial mixed models in DEseq2. RESULTS Time in physical contact with caregivers explained 10% of variation in beta diversity at 2 weeks' age. The number of caregivers in the first few weeks of life explained 9% of variation in beta diversity at 2 weeks and the number of individuals in physical contact at birth explained 11% of variation in beta diversity at 6 months. Skin-to-skin contact on the day of birth was positively associated with the abundance of eight genera. Infants held for by more individuals had greater abundance of eight genera. DISCUSSION Results reveal a potential mechanism (skin-to-skin and physical contact) by which caregivers influence the infant gastrointestinal microbiome. Our findings contribute to work exploring the social transmission of microbes.
Collapse
Affiliation(s)
- Kyle S Wiley
- Department of Anthropology, UCLA, Los Angeles, California, USA
- Department of Psychiatry & Biobehavioral Sciences, UCLA, Los Angeles, California, USA
| | - Andrew M Gregg
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Molly M Fox
- Department of Anthropology, UCLA, Los Angeles, California, USA
- Department of Psychiatry & Biobehavioral Sciences, UCLA, Los Angeles, California, USA
| | - Venu Lagishetty
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- UCLA Microbiome Center, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Curt A Sandman
- Department of Psychiatry and Human Behavior, UC Irvine, Irvine, California, USA
| | - Jonathan P Jacobs
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- UCLA Microbiome Center, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Laura M Glynn
- Department of Psychology, Chapman University, Orange, California, USA
| |
Collapse
|
26
|
Søegaard SH, Andersen MM, Rostgaard K, Davidsson OB, Olsen SF, Schmiegelow K, Hjalgrim H. Exclusive Breastfeeding Duration and Risk of Childhood Cancers. JAMA Netw Open 2024; 7:e243115. [PMID: 38530315 PMCID: PMC10966412 DOI: 10.1001/jamanetworkopen.2024.3115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/25/2024] [Indexed: 03/27/2024] Open
Abstract
Importance Breastfeeding has been suggested to protect against childhood cancers, particularly acute lymphoblastic leukemia (ALL). However, the evidence stems from case-control studies alone. Objective To investigate whether longer duration of exclusive breastfeeding is associated with decreased risk of childhood ALL and other childhood cancers. Design, Setting, and Participants This population-based cohort study used administrative data on exclusive breastfeeding duration from the Danish National Child Health Register. All children born in Denmark between January 2005 and December 2018 with available information on duration of exclusive breastfeeding were included. Children were followed up from age 1 year until childhood cancer diagnosis, loss to follow-up or emigration, death, age 15 years, or December 31, 2020. Data were analyzed from March to October 2023. Exposure Duration of exclusive breastfeeding in infancy. Main Outcomes and Measures Associations between duration of exclusive breastfeeding and risk of childhood cancer overall and by subtypes were estimated as adjusted hazard ratios (AHRs) with 95% CIs using stratified Cox proportional hazards regression models. Results A total of 309 473 children were included (51.3% boys). During 1 679 635 person-years of follow-up, 332 children (0.1%) were diagnosed with cancer at ages 1 to 14 years (mean [SD] age at diagnosis, 4.24 [2.67] years; 194 boys [58.4%]). Of these, 124 (37.3%) were diagnosed with hematologic cancers (81 [65.3%] were ALL, 74 [91.4%] of which were B-cell precursor [BCP] ALL), 44 (13.3%) with central nervous system tumors, 80 (24.1%) with solid tumors, and 84 (25.3%) with other and unspecified malignant neoplasms. Compared with exclusive breastfeeding duration of less than 3 months, exclusive breastfeeding for 3 months or longer was associated with a decreased risk of hematologic cancers (AHR, 0.66; 95% CI, 0.46-0.95), which was largely attributable to decreased risk of BCP-ALL (AHR, 0.62; 95% CI, 0.39-0.99), but not with risk of central nervous system tumors (AHR, 0.96; 95% CI, 0.51-1.88) or solid tumors (AHR, 0.87; 95% CI, 0.55-1.41). Conclusions and Relevance In this cohort study, longer duration of exclusive breastfeeding was associated with reduced risk of childhood BCP-ALL, corroborating results of previous case-control investigations in this field. To inform future preemptive interventions, continued research should focus on the potential biologic mechanisms underlying the observed association.
Collapse
Affiliation(s)
- Signe Holst Søegaard
- Danish Cancer Institute, Danish Cancer Society, Copenhagen, Denmark
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Mie Mølgaard Andersen
- Danish Cancer Institute, Danish Cancer Society, Copenhagen, Denmark
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Klaus Rostgaard
- Danish Cancer Institute, Danish Cancer Society, Copenhagen, Denmark
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Olafur Birgir Davidsson
- Danish Cancer Institute, Danish Cancer Society, Copenhagen, Denmark
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Sjurdur Frodi Olsen
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- University of the Faroe Islands, Torshavn, Faroe Islands
| | - Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
- Institute of Clinical Medicine, Faculty of Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Hjalgrim
- Danish Cancer Institute, Danish Cancer Society, Copenhagen, Denmark
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
- Institute of Clinical Medicine, Faculty of Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Hematology, University Hospital Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
27
|
Chawla D. Kangaroo Mother Care and Neonatal Skin Microbiome. Indian J Pediatr 2024; 91:213-214. [PMID: 38114861 DOI: 10.1007/s12098-023-04984-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 12/06/2023] [Indexed: 12/21/2023]
Affiliation(s)
- Deepak Chawla
- Department of Neonatology, Government Medical College Hospital, Chandigarh, India.
| |
Collapse
|
28
|
Lutpiatina L, Sulistyorini L, Yudhastuti R, Notobroto HB. Prediction of Toddlers Acute Respiratory Infection (ARI) to Become Pneumonia in Martapura Catchment Area, Banjar District, Indonesia. Glob Pediatr Health 2024; 11:2333794X241227694. [PMID: 38405016 PMCID: PMC10894530 DOI: 10.1177/2333794x241227694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 12/06/2023] [Accepted: 01/05/2024] [Indexed: 02/27/2024] Open
Abstract
Introduction. The pneumonia pattern in young children may vary across different catchment areas. Therefore, this study aims to analyze the predictive factors for toddlers with an Acute Respiratory Infection (ARI) developing into pneumonia in the catchment area of Banjar Regency, Indonesia. Methods. A case-control design, with 300 respondents, consisting of 106 cases and 194 controls. A questionnaire of interviews with mothers/caregivers of toddlers. Forty-one indicators data were analyzed using multiple logistic regression with backward stepwise regression to arrive at the final model. Results. The predictive factors for toddlers with pneumonia were the child's age (P-value .070), child development (P-value .007), breastfeeding (P-value .051), family income (P-value .026), and location of houses along the river (P-value .025). Conclusion. A prediction index for toddler pneumonia has been compiled, which can be applied to improve the health of lower middle-class toddlers requiring more government attention.
Collapse
Affiliation(s)
- Leka Lutpiatina
- Medical Laboratory Technology Poltekkes Kemenkes Banjarmasin, Banjarbaru, Indonesia
| | | | | | | |
Collapse
|
29
|
Lu D, Liu Y, Kang L, Zhang X, Hu J, Ye H, Huang B, Wu Y, Zhao J, Dai Z, Wang J, Han D. Maternal fiber-rich diet promotes early-life intestinal development in offspring through milk-derived extracellular vesicles carrying miR-146a-5p. J Nanobiotechnology 2024; 22:65. [PMID: 38365722 PMCID: PMC10870446 DOI: 10.1186/s12951-024-02344-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/12/2024] [Indexed: 02/18/2024] Open
Abstract
BACKGROUNDS The intestinal development in early life is profoundly influenced by multiple biological components of breast milk, in which milk-derived extracellular vesicles (mEVs) contain a large amount of vertically transmitted signal from the mother. However, little is known about how maternal fiber-rich diet regulates offspring intestinal development by influencing the mEVs. RESULTS In this study, we found that maternal resistant starch (RS) consumption during late gestation and lactation improved the growth and intestinal health of offspring. The mEVs in breast milk are the primary factor driving these beneficial effects, especially enhancing intestinal cell proliferation and migration. To be specific, administration of mEVs after maternal RS intake enhanced intestinal cell proliferation and migration in vivo (performed in mice model and indicated by intestinal histological observation, EdU assay, and the quantification of cyclin proteins) and in vitro (indicated by CCK8, MTT, EdU, and wound healing experiments). Noteworthily, miR-146a-5p was found to be highly expressed in the mEVs from maternal RS group, which also promotes intestinal cell proliferation in cells and mice models. Mechanically, miR-146a-5p target to silence the expression of ubiquitin ligase 3 gene NEDD4L, thereby inhibiting DVL2 ubiquitination, activating the Wnt pathway, and promoting intestinal development. CONCLUSION These findings demonstrated the beneficial role of mEVs in the connection between maternal fiber rich diet and offspring intestinal growth. In addition, we identified a novel miRNA-146a-5p-NEDD4L-β-catenin/Wnt signaling axis in regulating early intestinal development. This work provided a new perspective for studying the influence of maternal diet on offspring development.
Collapse
Affiliation(s)
- Dongdong Lu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
- Adaptation Physiology Group, Wageningen University & Research, Wageningen, 6700 AH, The Netherlands
| | - Yisi Liu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Luyuan Kang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Xiangyu Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jie Hu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Hao Ye
- Adaptation Physiology Group, Wageningen University & Research, Wageningen, 6700 AH, The Netherlands
| | - Bingxu Huang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yujun Wu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jinbiao Zhao
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Zhaolai Dai
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Junjun Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Dandan Han
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
30
|
Liu F, Tol AJ, Kuipers F, Oosterveer MH, van der Beek EM, van Leeuwen SS. Characterization of milk oligosaccharide and sialic acid content and their influence on brain sialic acid in a lean mouse model for gestational diabetes. Heliyon 2024; 10:e24539. [PMID: 38317966 PMCID: PMC10839809 DOI: 10.1016/j.heliyon.2024.e24539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 01/01/2024] [Accepted: 01/10/2024] [Indexed: 02/07/2024] Open
Abstract
Oligosaccharides and sialic acids (Sia) are bioactive components in milk that contribute to newborn development and health. Hyperglycemia in pregnancy (HIP) can have adverse effects on both mother and infant. HIP is associated with low-grade systemic inflammation. Inflammation influenced glycan composition, particularly of Sia-containing structures. We hypothesize that HIP and high-fat diet influence milk oligosaccharide composition, particularly sialylated oligosaccharides. Furthermore, we propose that milk Sia content influences pup brain Sia content. To test these hypotheses we (i) characterize mouse milk oligosaccharides and Sia concentrations in mouse milk of a GDM mouse model with dietary fat intake intervention; and (ii) determine Sia levels in offspring brains. The concentrations of oligosaccharides and Sia in mouse milk and offspring's brains were quantified using UPLC-FLD analysis. Analyses were performed on surplus samples from a previous study, where HIP was induced by combining high-fat diet (HF) feeding and low-dose streptozotocin injections in C57Bl/6NTac female mice. The previous study described the metabolic effects of HIP on dams and offspring. We detected 21 mouse milk oligosaccharides, including 9 neutral and 12 acidic structures using UPLC-MS. A total of 8 structures could be quantified using UPLC-FLD. Maternal HIP and HF diet during lactation influenced sialylated oligosaccharide concentrations in mouse milk and total and free sialic acid concentrations. Sia content in offspring brain was associated with total and free Neu5Gc in mouse milk of dams, but no correlations with HIP or maternal diet were observed.
Collapse
Affiliation(s)
- Fan Liu
- Laboratory of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Angela J.C. Tol
- Laboratory of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Folkert Kuipers
- Laboratory of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| | - Maaike H. Oosterveer
- Laboratory of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| | - Eline M. van der Beek
- Laboratory of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Sander S. van Leeuwen
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| |
Collapse
|
31
|
Xu D, Zhou S, Liu Y, Scott AL, Yang J, Wan F. Complement in breast milk modifies offspring gut microbiota to promote infant health. Cell 2024; 187:750-763.e20. [PMID: 38242132 PMCID: PMC10872564 DOI: 10.1016/j.cell.2023.12.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/03/2023] [Accepted: 12/14/2023] [Indexed: 01/21/2024]
Abstract
Breastfeeding offers demonstrable benefits to newborns and infants by providing nourishment and immune protection and by shaping the gut commensal microbiota. Although it has been appreciated for decades that breast milk contains complement components, the physiological relevance of complement in breast milk remains undefined. Here, we demonstrate that weanling mice fostered by complement-deficient dams rapidly succumb when exposed to murine pathogen Citrobacter rodentium (CR), whereas pups fostered on complement-containing milk from wild-type dams can tolerate CR challenge. The complement components in breast milk were shown to directly lyse specific members of gram-positive gut commensal microbiota via a C1-dependent, antibody-independent mechanism, resulting in the deposition of the membrane attack complex and subsequent bacterial lysis. By selectively eliminating members of the commensal gut community, complement components from breast milk shape neonate and infant gut microbial composition to be protective against environmental pathogens such as CR.
Collapse
Affiliation(s)
- Dongqing Xu
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Siyu Zhou
- NHC Key Laboratory of Systems Biology of Pathogens, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Yue Liu
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Alan L Scott
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA; Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Jian Yang
- NHC Key Laboratory of Systems Biology of Pathogens, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Fengyi Wan
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA; Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
32
|
Ennis D, Shmorak S, Jantscher-Krenn E, Yassour M. Longitudinal quantification of Bifidobacterium longum subsp. infantis reveals late colonization in the infant gut independent of maternal milk HMO composition. Nat Commun 2024; 15:894. [PMID: 38291346 PMCID: PMC10827747 DOI: 10.1038/s41467-024-45209-y] [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: 07/19/2023] [Accepted: 01/15/2024] [Indexed: 02/01/2024] Open
Abstract
Breast milk contains human milk oligosaccharides (HMOs) that cannot be digested by infants, yet nourish their developing gut microbiome. While Bifidobacterium are the best-known utilizers of individual HMOs, a longitudinal study examining the evolving microbial community at high-resolution coupled with mothers' milk HMO composition is lacking. Here, we developed a high-throughput method to quantify Bifidobacterium longum subsp. infantis (BL. infantis), a proficient HMO-utilizer, and applied it to a longitudinal cohort consisting of 21 mother-infant dyads. We observed substantial changes in the infant gut microbiome over the course of several months, while the HMO composition in mothers' milk remained relatively stable. Although Bifidobacterium species significantly influenced sample variation, no specific HMOs correlated with Bifidobacterium species abundance. Surprisingly, we found that BL. infantis colonization began late in the breastfeeding period both in our cohort and in other geographic locations, highlighting the importance of focusing on BL. infantis dynamics in the infant gut.
Collapse
Affiliation(s)
- Dena Ennis
- Microbiology & Molecular Genetics Department, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shimrit Shmorak
- Microbiology & Molecular Genetics Department, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | | | - Moran Yassour
- Microbiology & Molecular Genetics Department, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
- The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
33
|
Allert M, Ferretti P, Johnson KE, Heisel T, Gonia S, Knights D, Fields DA, Albert FW, Demerath EW, Gale CA, Blekhman R. Assembly, stability, and dynamics of the infant gut microbiome are linked to bacterial strains and functions in mother's milk. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.28.577594. [PMID: 38328166 PMCID: PMC10849666 DOI: 10.1101/2024.01.28.577594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
The establishment of the gut microbiome in early life is critical for healthy infant development. Although human milk is recommended as the sole source of nutrition for the human infant, little is known about how variation in milk composition, and especially the milk microbiome, shapes the microbial communities in the infant gut. Here, we quantified the similarity between the maternal milk and the infant gut microbiome using 507 metagenomic samples collected from 195 mother-infant pairs at one, three, and six months postpartum. We found that the microbial taxonomic overlap between milk and the infant gut was driven by bifidobacteria, in particular by B. longum. Infant stool samples dominated by B. longum also showed higher temporal stability compared to samples dominated by other species. We identified two instances of strain sharing between maternal milk and the infant gut, one involving a commensal (B. longum) and one a pathobiont (K. pneumoniae). In addition, strain sharing between unrelated infants was higher among infants born at the same hospital compared to infants born in different hospitals, suggesting a potential role of the hospital environment in shaping the infant gut microbiome composition. The infant gut microbiome at one month compared to six months of age was enriched in metabolic pathways associated with de-novo molecule biosynthesis, suggesting that early colonisers might be more versatile and metabolically independent compared to later colonizers. Lastly, we found a significant overlap in antimicrobial resistance genes carriage between the mother's milk and their infant's gut microbiome. Taken together, our results suggest that the human milk microbiome has an important role in the assembly, composition, and stability of the infant gut microbiome.
Collapse
Affiliation(s)
- Mattea Allert
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| | - Pamela Ferretti
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Kelsey E Johnson
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| | - Timothy Heisel
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Sara Gonia
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Dan Knights
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, USA
- BioTechnology Institute, College of Biological Sciences, University of Minnesota, Minneapolis, MN, USA
| | - David A Fields
- Department of Pediatrics, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Frank W Albert
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| | - Ellen W Demerath
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| | - Cheryl A Gale
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Ran Blekhman
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| |
Collapse
|
34
|
Sun W, Tao L, Qian C, Xue P, Tong X, Yang L, Lu F, Wan H, Tao Y. Human milk oligosaccharides and the association with microbiota in colostrum: a pilot study. Arch Microbiol 2024; 206:58. [PMID: 38191870 PMCID: PMC10774193 DOI: 10.1007/s00203-023-03787-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 10/25/2023] [Accepted: 12/04/2023] [Indexed: 01/10/2024]
Abstract
HMOs (Human milk oligosaccharide) has an impact on maternal and infant health. Colostrum samples of 70 breastfeeding women in China were collected and recorded clinical characteristics. The major oligosaccharides and microbiota were quantitated in colostrum. The concentration of fucosylated HMOs in primipara was higher than that of multipara (p = 0.030). The concentration of N-acetylated HMOs in vaginal delivery milk was less than that of cesarean (p = 0.038). Non-fucosylated HMOs of breastfeeding women were less than that of breast pump (p = 0.038). Meanwhile, the concentration of LNT was positively correlated with Lactobacillus (r = 0.250, p = 0.037). DS-LNT was negatively correlated with Staphylococcus (r = - 0.240, p = 0.045). There was a positive correlation of Streptococcus with LNFP II (r = 0.314, p = 0.011) and 3-SL (r = 0.322, p = 0.009). In addition, there was a negative correlation between 2'-FL and 3-FL (r = - 0.465, p = 0.001). There was a positive correlation between LNT and LNnT (r = 0.778, p = 0.001). Therefore, the concentration of HMOs is related to number of deliveries, delivery mode, lactation mode and perinatal antibiotic. The concentration of HMOs is related to Lactobacillus, Streptococcus and Streptococcus in colostrum. In addition, there are connections between different oligosaccharides in content. The study protocol was also registered in the ClinicalTrails.gov (ChiCTR2200064454) (Oct. 2022).
Collapse
Affiliation(s)
- Wen Sun
- Department of Traditional Chinese Medicine, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, 200434, China
| | - Lin Tao
- Department of Traditional Chinese Medicine, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, 200434, China
| | - Chen Qian
- Department of Traditional Chinese Medicine, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, 200434, China
| | - Peipei Xue
- Department of Traditional Chinese Medicine, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, 200434, China
| | - Xiankun Tong
- Laboratory of Immunopharmacology, Shanghai Institute of Materia Medical, Chinese Academy of Sciences, Shanghai, 201210, China
| | - Li Yang
- Laboratory of Immunopharmacology, Shanghai Institute of Materia Medical, Chinese Academy of Sciences, Shanghai, 201210, China
| | - Fang Lu
- Department of Gynaecology and Obstetrics, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, 200434, China
| | - Hua Wan
- Department of Breast, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Yingna Tao
- Department of Traditional Chinese Medicine, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, 200434, China.
| |
Collapse
|
35
|
Shi B, Li H, He X. Advancing lifelong precision medicine for cardiovascular diseases through gut microbiota modulation. Gut Microbes 2024; 16:2323237. [PMID: 38411391 PMCID: PMC10900281 DOI: 10.1080/19490976.2024.2323237] [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: 12/19/2023] [Accepted: 02/21/2024] [Indexed: 02/28/2024] Open
Abstract
The gut microbiome is known as the tenth system of the human body that plays a vital role in the intersection between health and disease. The considerable inter-individual variability in gut microbiota poses both challenges and great prospects in promoting precision medicine in cardiovascular diseases (CVDs). In this review, based on the development, evolution, and influencing factors of gut microbiota in a full life circle, we summarized the recent advances on the characteristic alteration in gut microbiota in CVDs throughout different life stages, and depicted their pathological links in mechanism, as well as the highlight achievements of targeting gut microbiota in CVDs prevention, diagnosis and treatment. Personalized strategies could be tailored according to gut microbiota characteristics in different life stages, including gut microbiota-blood metabolites combined prediction and diagnosis, dietary interventions, lifestyle improvements, probiotic or prebiotic supplements. However, to fulfill the promise of a lifelong cardiovascular health, more mechanism studies should progress from correlation to causality and decipher novel mechanisms linking specific microbes and CVDs. It is also promising to use the burgeoning artificial intelligence and machine learning to target gut microbiota for developing diagnosis system and screening for new therapeutic interventions.
Collapse
Affiliation(s)
- Bozhong Shi
- Department of Cardiothoracic Surgery, Shanghai Children’s Medical Center Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haoyu Li
- Department of Cardiothoracic Surgery, Shanghai Children’s Medical Center Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaomin He
- Department of Cardiothoracic Surgery, Shanghai Children’s Medical Center Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children’s Medical Center, National Children’s Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
36
|
Vahkal B, Altosaar I, Tremblay E, Gagné D, Hüttman N, Minic Z, Côté M, Blais A, Beaulieu J, Ferretti E. Gestational age at birth influences protein and RNA content in human milk extracellular vesicles. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e128. [PMID: 38938674 PMCID: PMC11080785 DOI: 10.1002/jex2.128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/26/2023] [Accepted: 11/02/2023] [Indexed: 06/29/2024]
Abstract
Human milk extracellular vesicles (HM EVs) are proposed to protect against disease development in infants. This protection could in part be facilitated by the bioactive EV cargo of proteins and RNA. Notably, mothers birth infants of different gestational ages with unique needs, wherein the EV cargo of HM may diverge. We collected HM from lactating mothers within two weeks of a term or preterm birth. Following purification of EVs, proteins and mRNA were extracted for proteomics and sequencing analyses, respectively. Over 2000 protein groups were identified, and over 8000 genes were quantified. The total number of proteins and mRNA did not differ significantly between the two conditions, while functional bioinformatics of differentially expressed cargo indicated enrichment in immunoregulatory cargo for preterm HM EVs. In term HM EVs, significantly upregulated cargo was enriched in metabolism-related functions. Based on gene expression signatures from HM-contained single cell sequencing data, we proposed that a larger portion of preterm HM EVs are secreted by immune cells, whereas term HM EVs contain more signatures of lactocyte epithelial cells. Proposed differences in EV cargo could indicate variation in mother's milk based on infants' gestational age and provide basis for further functional characterisation.
Collapse
Affiliation(s)
- Brett Vahkal
- Department of Biochemistry, Microbiology and ImmunologyUniversity of OttawaOttawaCanada
- uOttawa Centre for Infection, Immunity, and InflammationOttawaCanada
- Ottawa Institute of Systems BiologyOttawaCanada
| | - Illimar Altosaar
- Department of Biochemistry, Microbiology and ImmunologyUniversity of OttawaOttawaCanada
| | - Eric Tremblay
- Department of Immunology and Cell BiologyUniversité de SherbrookeSherbrookeCanada
| | - David Gagné
- Department of Immunology and Cell BiologyUniversité de SherbrookeSherbrookeCanada
| | - Nico Hüttman
- Faculty of Science, John L. Holmes Mass Spectrometry FacilityUniversity of OttawaOttawaCanada
| | - Zoran Minic
- Faculty of Science, John L. Holmes Mass Spectrometry FacilityUniversity of OttawaOttawaCanada
| | - Marceline Côté
- Department of Biochemistry, Microbiology and ImmunologyUniversity of OttawaOttawaCanada
- uOttawa Centre for Infection, Immunity, and InflammationOttawaCanada
- Ottawa Institute of Systems BiologyOttawaCanada
| | - Alexandre Blais
- Department of Biochemistry, Microbiology and ImmunologyUniversity of OttawaOttawaCanada
- uOttawa Centre for Infection, Immunity, and InflammationOttawaCanada
- Ottawa Institute of Systems BiologyOttawaCanada
- Brain and Mind InstituteUniversity of OttawaOttawaCanada
- Éric Poulin Centre for Neuromuscular DiseaseOttawaCanada
| | | | - Emanuela Ferretti
- Department of Pediatrics, Division of NeonatologyChildren's Hospital of Eastern OntarioOttawaCanada
| |
Collapse
|
37
|
Wang H, Li QF, Xu XF, Hu XL. Effects of Sublingual Colostrum Application on Oral and Intestinal Flora of Extremely Low Birth Weight Infants. Endocr Metab Immune Disord Drug Targets 2024; 24:489-494. [PMID: 37711000 DOI: 10.2174/1871530323666230913105820] [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: 02/13/2023] [Revised: 07/26/2023] [Accepted: 07/28/2023] [Indexed: 09/16/2023]
Abstract
BACKGROUND The aim of this study is to analyze the effects of colostrum application on the establishment of normal flora in the intestinal tracts and oral cavities of extremely low birth weight infants (ELBWI). METHODS A prospective cohort study design was adopted following the STROBE guidelines (Supplementary File 1). Colostrum was administered immediately after obtaining maternal breast milk using a special sterile cotton swab. There were no specific treatments for infants who did not receive colostrum. This experiment was completed on day 5 post-birth and the patients were divided into the colostrum and control groups, corresponding to whether or not colostrum was administered. Throat swabs and stool samples were collected on days 1 and 5 post-birth. RESULTS Using the conventional bacteria cultivation technique, the detection rate of bacteria in 98 cases of meconium at birth was 15.31%. On day 5, the detection rates of Staphylococcus in the colostrum and control groups were 36.54% and 34.78%, with no significant difference between them (P = 0.856), and that of Enterococcus was 26.92% and 13.04%, respectively, with no statistically significant difference (P = 0.089). Likewise, at birth, the detection rate of bacteria in 98 cases of throat swabs was 27.55%. On day 5, the detection rate of Streptococcus in the colostrum and control groups was 78.85% and 50.00%, respectively, recording a statistically significant difference this time (P = 0.003). CONCLUSION Colostrum application had limited effects on intestinal flora colonization but contributes to physiological oral flora colonization.
Collapse
Affiliation(s)
- Hua Wang
- Department of NICU, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Qiu-Fang Li
- Department of nursing, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Xin-Fen Xu
- Department of nursing, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Xiao-Li Hu
- Department of obstetrics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| |
Collapse
|
38
|
Sun Y, Zhang S, He H, Chen H, Nie Q, Li S, Cheng J, Zhang B, Zheng Z, Pan S, Huang P, Lian L, Hu J, Nie S. Comprehensive evaluation of the prebiotic properties of Dendrobium officinale polysaccharides, β-glucan, and inulin during in vitro fermentation via multi-omics analysis. Int J Biol Macromol 2023; 253:127326. [PMID: 37820907 DOI: 10.1016/j.ijbiomac.2023.127326] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 10/02/2023] [Accepted: 10/06/2023] [Indexed: 10/13/2023]
Abstract
Dietary fiber is crucial for human health mainly due to its impact on gut microbiota structure and metabolites. This study aimed to investigate the impact of Dendrobium officinale polysaccharides (DOP) and two common fibers (β-glucan and inulin) on the gut microbiome structure and metabolic profile in vitro. Fecal samples were obtained from 30 healthy volunteers, which were then individually subjected to fermentation with each type of fiber. The results revealed that all fibers were efficiently degraded by gut microbiota, with DOP exhibiting a slower fermentation rate compared to β-glucan and inulin. The fermentation of all fibers led to a significant increase in the production of short-chain fatty acids (SCFAs) and a reduction in branched-chain fatty acids (BCFAs), sulfides, phenols, and indole. Moreover, the abundance of unclassified Enterobacteriaceae, which was positively correlated with sulfide, phenols, and indole levels, was significantly reduced by all fibers. Additionally, DOP specifically promoted the growth of Parabacteroides, while β-glucan and inulin promoted the growth of Bifidobacterium and Faecalibacterium. Taken together, these findings enhance our understanding of the role of DOP, β-glucan, and inulin in modulating gut microbiota and metabolites, where the fermentation with fecal bacteria from different volunteers could provide valuable insights for personalized therapeutic approaches.
Collapse
Affiliation(s)
- Yonggan Sun
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, Nanchang 330047, China
| | - Shanshan Zhang
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, Nanchang 330047, China
| | - Huijun He
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, Nanchang 330047, China
| | - Haihong Chen
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, Nanchang 330047, China
| | - Qixing Nie
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, Nanchang 330047, China
| | - Song Li
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, Nanchang 330047, China
| | - Jiaobo Cheng
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, Nanchang 330047, China
| | - Baojie Zhang
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, Nanchang 330047, China
| | - Zhitian Zheng
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, Nanchang 330047, China
| | - Shijie Pan
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, Nanchang 330047, China
| | - Ping Huang
- Department of Nutrition, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Lu Lian
- Department of Nutrition, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Jielun Hu
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, Nanchang 330047, China
| | - Shaoping Nie
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, Nanchang 330047, China.
| |
Collapse
|
39
|
Huo J, Zhang R, Wu X, Fu C, Hu J, Hu X, Sun W, Chen Z, Zhu X. Active polypeptide MDANP protect against necrotizing enterocolitis (NEC) by regulating the PERK-eIF2ɑ-QRICH1 axis. Sci Rep 2023; 13:22912. [PMID: 38129421 PMCID: PMC10739873 DOI: 10.1038/s41598-023-44194-4] [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: 03/23/2023] [Accepted: 10/04/2023] [Indexed: 12/23/2023] Open
Abstract
The effect of MDANP effects on ER stress signalling not well known or elucidated. Endoplasmic reticulum (ER) stress plays a critical role in necrotizing enterocolitis (NEC) pathogenesis through the PERK-eIF2ɑ-QRICH1 axis. The present study aimed to explore the protective effects of MDANP in NEC development. Firstly, a function screening was designed to identify the candidate peptides in human milk, and then the identified peptides were validated in NEC patients. In vivo, NEC was induced in mice pups and divided into four groups: (1) control group, (2) NEC group, (3) MDANP + NEC group, and (4) NS + NEC group. In vitro, lentivirus-mediated QRICH1 silencing, was used to transfect NCM460 cell lines, then stimulated with LPS. After LPS stimulation, cells were treated with chemically synthesized MDANP, and the essential proteins in the QRICH1 signalling pathway in cells were tested and compared. After the small-scale screening, a peptide (SKSKKFRRPDIQYPDATDED) named MDANP was determined as the principal peptide. Its protective effect against NEC through inhibiting the expression of ERS key proteins and impeding the intestinal cells' apoptosis was observed in the animal models. Furthermore, the inhibitive effect of MDANP on apoptosis of intestinal epithelial cells through modulating the PERK-eIF2ɑ-QRICH1 ERS pathway was also confirmed in vitro. Taken together, our data suggest that MDANP effectively ameliorates apoptosis in NEC through attenuating PERK-eIF2ɑ-QRICH1.
Collapse
Affiliation(s)
- Jie Huo
- Department of Neonatology, Children's Hospital of Soochow University, No. 92 Zhongnan Street, Industrial Park, Suzhou, Jiangsu, 215025, People's Republic of China
- Department of Neonatology, Yangzhou Maternity and Child Health Care Hospital, Yangzhou, People's Republic of China
| | - Rui Zhang
- Department of Neonatology, Children's Hospital of Soochow University, No. 92 Zhongnan Street, Industrial Park, Suzhou, Jiangsu, 215025, People's Republic of China
| | - Xinping Wu
- Department of Neonatology, Yangzhou Maternity and Child Health Care Hospital, Yangzhou, People's Republic of China
| | - Changchang Fu
- Department of Neonatology, Children's Hospital of Soochow University, No. 92 Zhongnan Street, Industrial Park, Suzhou, Jiangsu, 215025, People's Republic of China
| | - Jinhui Hu
- Department of Neonatology, Children's Hospital of Soochow University, No. 92 Zhongnan Street, Industrial Park, Suzhou, Jiangsu, 215025, People's Republic of China
- Neonatal Medical Center, Huai'an Maternity and Child Health Care Hospital, Xuzhou Medical University, Huai'an, People's Republic of China
| | - Xiaohan Hu
- Department of Neonatology, Children's Hospital of Soochow University, No. 92 Zhongnan Street, Industrial Park, Suzhou, Jiangsu, 215025, People's Republic of China
| | - Wenqiang Sun
- Department of Neonatology, Children's Hospital of Soochow University, No. 92 Zhongnan Street, Industrial Park, Suzhou, Jiangsu, 215025, People's Republic of China
| | - Zhenjiang Chen
- Department of Neonatology, Children's Hospital of Soochow University, No. 92 Zhongnan Street, Industrial Park, Suzhou, Jiangsu, 215025, People's Republic of China
| | - Xueping Zhu
- Department of Neonatology, Children's Hospital of Soochow University, No. 92 Zhongnan Street, Industrial Park, Suzhou, Jiangsu, 215025, People's Republic of China.
| |
Collapse
|
40
|
Zwierz A, Domagalski K, Masna K, Walentowicz P, Burduk P. Impact of Breastfeeding Duration on Adenoid Hypertrophy, Snoring and Acute Otitis Media: A Case-Control Study in Preschool Children. J Clin Med 2023; 12:7683. [PMID: 38137751 PMCID: PMC10743536 DOI: 10.3390/jcm12247683] [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: 11/02/2023] [Revised: 12/03/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND The aim of this study was to analyze the relationship between breastfeeding duration and adenoid size, snoring and acute otitis media (AOM). METHODS We analyzed the medical history of children admitted to the ENT outpatient clinic in 2022 and 2023, reported symptoms, ear, nose and throat (ENT) examination, and flexible nasopharyngoscopy examination of 145 children aged 3-5 years. RESULTS Breastfeeding duration of 3 and 6 months or more had a significant effect on the reduction of snoring (p = 0.021; p = 0.039). However, it had no effect on the adenoid size, mucus coverage and sleeping with an open mouth. Snoring was correlated with open mouth sleeping (p < 0.001), adenoid size with a 75% A/C ratio or more (p < 0.001), and adenoid mucus coverage in the Mucus of Adenoid Scale by Nasopharyngoscopy Assessment-MASNA scale (p = 0.009). Children who were breastfed for less than 3 months had more than a four-fold greater risk of snoring. There was a statistically significant correlation between AOM and gender (p = 0.033), breastfeeding duration in groups fed 1, 3 or 6 months or more (p = 0.018; p = 0.004; p = 0.004) and those fed with mother's breast milk 3 or 6 months or more (p = 0.009; p = 0.010). Moreover, a correlation was found between adenoid size and mucus coverage, tympanogram, and open-mouth sleeping (p < 0.001). Independent factors of snoring in 3- to 5-year-old children were breastfeeding duration of less than 3 months (p = 0.032), adenoid size with an A/C ratio of 75% or more (p = 0.023) and open mouth sleeping (p = 0.001). CONCLUSIONS Children breastfed for 3 and 6 months or more exhibited reduced rates of snoring. There was no effect of breastfeeding duration on adenoid size in children aged 3 to 5 years, suggesting that the link between breastfeeding duration and snoring is primarily associated with craniofacial development and muscle tone stimulation. A breastfeeding duration of 1 month or more plays a key role in reducing the rate of AOM. The mother's milk plays a protective role against AOM. The presence of mucus might be responsible for snoring in preschool children. A medical history of breastfeeding should be taken into consideration when snoring children are suspected of adenoid hypertrophy.
Collapse
Affiliation(s)
- Aleksander Zwierz
- Department of Otolaryngology, Phoniatrics and Audiology, Faculty of Health Sciences, Ludwik Rydygier Collegium Medicum, Nicolaus Copernicus University, 75 Ujejskiego Street, 85-168 Bydgoszcz, Poland; (K.M.); (P.B.)
| | - Krzysztof Domagalski
- Department of Immunology, Faculty of Biological and Veterinary Sciences, Nicolaus Copenicus University, 87-100 Toruń, Poland;
| | - Krystyna Masna
- Department of Otolaryngology, Phoniatrics and Audiology, Faculty of Health Sciences, Ludwik Rydygier Collegium Medicum, Nicolaus Copernicus University, 75 Ujejskiego Street, 85-168 Bydgoszcz, Poland; (K.M.); (P.B.)
| | - Paweł Walentowicz
- Department of Obstetrics, Gynecology and Gynecologic Oncology, Regional Polyclinical Hospital, 87-100 Toruń, Poland;
| | - Paweł Burduk
- Department of Otolaryngology, Phoniatrics and Audiology, Faculty of Health Sciences, Ludwik Rydygier Collegium Medicum, Nicolaus Copernicus University, 75 Ujejskiego Street, 85-168 Bydgoszcz, Poland; (K.M.); (P.B.)
| |
Collapse
|
41
|
Mun D, Kang M, Shin M, Choi HJ, Kang AN, Ryu S, Unno T, Maburutse BE, Oh S, Kim Y. Alleviation of DSS-induced colitis via bovine colostrum-derived extracellular vesicles with microRNA let-7a-5p is mediated by regulating Akkermansia and β-hydroxybutyrate in gut environments. Microbiol Spectr 2023; 11:e0012123. [PMID: 37966243 PMCID: PMC10714758 DOI: 10.1128/spectrum.00121-23] [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: 01/09/2023] [Accepted: 10/11/2023] [Indexed: 11/16/2023] Open
Abstract
IMPORTANCE Even though studying on the possible involvement of extracellular vesicles (EVs) in host-microbe interactions, how these relationships mediate host physiology has not clarified yet. Our current findings provide insights into the encouraging benefits of dietary source-derived EVs and microRNAs (miRNAs) on organic acid production and ultimately stimulating gut microbiome for human health, suggesting that supplementation of dietary colostrum EVs and miRNAs is a novel preventive strategy for the treatment of inflammatory bowel disease.
Collapse
Affiliation(s)
- Daye Mun
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, South Korea
| | - Minkyoung Kang
- Department of Functional Food and Biotechnology, Jeonju University, Jeonju, South Korea
| | - Minhye Shin
- Department of Microbiology, College of Medicine, Inha University, Incheon, South Korea
| | - Hye Jin Choi
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, South Korea
| | - An Na Kang
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, South Korea
| | - Sangdon Ryu
- Division of Evironmental Meterials, Honam National Institute of Biological Resources, Mokpo, South Korea
| | - Tatsuya Unno
- Department of Microbiology, Chungbuk National University, Cheongju, South Korea
| | - Brighton E. Maburutse
- Department of Animal Production Sciences, Marondera University of Agricultural Sciences & Technology, Marondera, Zimbabwe
| | - Sangnam Oh
- Department of Functional Food and Biotechnology, Jeonju University, Jeonju, South Korea
| | - Younghoon Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, South Korea
| |
Collapse
|
42
|
Froń A, Orczyk-Pawiłowicz M. Understanding the Immunological Quality of Breast Milk in Maternal Overweight and Obesity. Nutrients 2023; 15:5016. [PMID: 38140275 PMCID: PMC10746120 DOI: 10.3390/nu15245016] [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: 11/14/2023] [Revised: 11/30/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
Maternal obesity, affecting many pregnant women globally, not only poses immediate health risks but also modulates breast milk composition. Obesity is linked to inflammation and oxidative stress, impacting breast milk's immune properties. This paper explores the intricate relationship between maternal metabolic disorders, such as obesity, and breast milk's immunological components. We conducted a thorough search for original and review articles published until 17 October 2023 in the PUBMED/Scopus database. This search included several terms related to human breast milk, immunological properties, and obesity. Articles were selected with the consensus of all authors. Maternal metabolic disorders have discernible effects on the composition of immune-related components in breast milk, such as immunoglobulins, lactoferrin, leptin, ghrelin, adiponectin, C-reactive protein, growth factors, extracellular vesicles, and lymphocytes. These changes in breast milk composition can significantly impact the newborn's immune system, with potential long-term health implications beyond the immediate postnatal period. Maternal metabolic health is a critical factor in shaping the health trajectory of the neonate through breastfeeding, although the full advantages of breastfeeding for children of mothers with obesity remain uncertain. Ongoing research aims to understand and unravel these links.
Collapse
Affiliation(s)
- Anita Froń
- Division of Chemistry and Immunochemistry, Department of Biochemistry and Immunochemistry, Wroclaw Medical University, M. Skłodowskiej-Curie 48/50, 50-369 Wroclaw, Poland
| | - Magdalena Orczyk-Pawiłowicz
- Division of Chemistry and Immunochemistry, Department of Biochemistry and Immunochemistry, Wroclaw Medical University, M. Skłodowskiej-Curie 48/50, 50-369 Wroclaw, Poland
| |
Collapse
|
43
|
Wong PY, Yip C, Lemberg DA, Day AS, Leach ST. Evolution of a Pathogenic Microbiome. J Clin Med 2023; 12:7184. [PMID: 38002796 PMCID: PMC10672640 DOI: 10.3390/jcm12227184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/16/2023] [Accepted: 11/16/2023] [Indexed: 11/26/2023] Open
Abstract
The process of microbiome development arguably begins before birth. Vertical transmission of bacteria from the mother to the infant is a keystone event in microbiome development. Subsequent to birth, the developing microbiome is vulnerable to influence from a wide range of factors. Additionally, the microbiome can influence the health and development of the host infant. This intricate interaction of the gastrointestinal microbiome and the host has been described as both symbiotic and dysbiotic. Defining these terms, a symbiotic microbiome is where the microbiome and host provide mutual benefit to each other. A pathogenic microbiome, or more precisely a gastrointestinal microbiome associated with disease, is increasing described as dysbiotic. This review seeks to investigate the factors that contribute to evolving a disease-causing or 'dysbiotic' microbiome. This review covers the development of the gastrointestinal microbiome in infants, the interaction of the microbiome with the host, and its contribution to host immunity and investigates specific features of the gastrointestinal microbiome that are associated with disease.
Collapse
Affiliation(s)
- Pui Yin Wong
- Discipline of Paediatrics, School of Clinical Medicine, University of NSW, Sydney 2052, Australia; (P.Y.W.); (C.Y.); (A.S.D.)
| | - Carmen Yip
- Discipline of Paediatrics, School of Clinical Medicine, University of NSW, Sydney 2052, Australia; (P.Y.W.); (C.Y.); (A.S.D.)
| | - Daniel A. Lemberg
- Department of Gastroenterology, Sydney Children’s Hospital Randwick, Sydney 2031, Australia;
| | - Andrew S. Day
- Discipline of Paediatrics, School of Clinical Medicine, University of NSW, Sydney 2052, Australia; (P.Y.W.); (C.Y.); (A.S.D.)
- Department of Gastroenterology, Sydney Children’s Hospital Randwick, Sydney 2031, Australia;
- Department of Paediatrics, University of Otago, Christchurch 8011, New Zealand
| | - Steven T. Leach
- Discipline of Paediatrics, School of Clinical Medicine, University of NSW, Sydney 2052, Australia; (P.Y.W.); (C.Y.); (A.S.D.)
| |
Collapse
|
44
|
Gómez-Ferrer M, Amaro-Prellezo E, Albiach-Delgado A, Ten-Domenech I, Kuligowski J, Sepúlveda P. Identification of omega-3 oxylipins in human milk-derived extracellular vesicles with pro-resolutive actions in gastrointestinal inflammation. Front Immunol 2023; 14:1293737. [PMID: 38054009 PMCID: PMC10694275 DOI: 10.3389/fimmu.2023.1293737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 10/27/2023] [Indexed: 12/07/2023] Open
Abstract
Introduction Premature infants (PIs) are at risk of suffering necrotizing enterocolitis (NEC), and infants consuming human milk (HM) show a lower incidence than infants receiving formula. The composition of HM has been studied in depth, but the lipid content of HM-derived small extracellular vesicles (HM sEVs) remains unexplored. Identifying these molecules and their biological effects has potential for the treatment of intestinal disorders in PIs and could contribute to the development of HM-based fortified formulas. Methods We isolated HM sEVs from HM samples and analyzed their oxylipin content using liquid chromatography coupled to mass spectrometry, which revealed the presence of anti-inflammatory oxylipins. We then examined the efficacy of a mixture of these oxylipins in combating inflammation and fibrosis, in vitro and in a murine model of inflammatory bowel disease (IBD). Results HM-related sEVs contained higher concentrations of oxylipins derived from docosahexaenoic acid, an omega-3 fatty acid. Three anti-inflammatory oxylipins, 14-HDHA, 17-HDHA, and 19,20-DiHDPA (ω3 OXLP), demonstrated similar efficacy to HM sEVs in preventing cell injury, inducing re-epithelialization, mitigating fibrosis, and modulating immune responses. Both ω3 OXLP and HM sEVs effectively reduced inflammation in IBD-model mice, preventing colon shortening, infiltration of inflammatory cells and tissue fibrosis. Discussion Incorporating this unique cocktail of oxylipins into fortified milk formulas might reduce the risk of NEC in PIs and also provide immunological and neurodevelopmental support.
Collapse
Affiliation(s)
- Marta Gómez-Ferrer
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital La Fe, Valencia, Spain
| | - Elena Amaro-Prellezo
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital La Fe, Valencia, Spain
| | - Abel Albiach-Delgado
- Neonatal Research Group, Health Research Institute Hospital La Fe, Valencia, Spain
| | - Isabel Ten-Domenech
- Neonatal Research Group, Health Research Institute Hospital La Fe, Valencia, Spain
| | - Julia Kuligowski
- Neonatal Research Group, Health Research Institute Hospital La Fe, Valencia, Spain
| | - Pilar Sepúlveda
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital La Fe, Valencia, Spain
- Cardiology Service, Hospital Universitari i Politècnic La Fe, Valencia, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Carlos III Institute of Health, Madrid, Spain
- Department of Pathology, University of Valencia, Valencia, Spain
| |
Collapse
|
45
|
Zhu L, Li H, Luo T, Deng Z, Li J, Zheng L, Zhang B. Human Milk Oligosaccharides: A Critical Review on Structure, Preparation, Their Potential as a Food Bioactive Component, and Future Perspectives. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:15908-15925. [PMID: 37851533 DOI: 10.1021/acs.jafc.3c04412] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
Human milk is the gold standard for infant feeding. Human milk oligosaccharides (HMOs) are a unique group of oligosaccharides in human milk. Great interest in HMOs has grown in recent years due to their positive effects on various aspects of infant health. HMOs provide various physiologic functions, including establishing a balanced infant's gut microbiota, strengthening the gastrointestinal barrier, preventing infections, and potential support to the immune system. However, the clinical application of HMOs is challenging due to their specificity to human milk and the difficulties and high costs associated with their isolation and synthesis. Here, the differences in oligosaccharides in human and other mammalian milk are compared, and the synthetic strategies to access HMOs are summarized. Additionally, the potential use and molecular mechanisms of HMOs as a new food bioactive component in different diseases, such as infection, necrotizing enterocolitis, diabetes, and allergy, are critically reviewed. Finally, the current challenges and prospects of HMOs in basic research and application are discussed.
Collapse
Affiliation(s)
- Liuying Zhu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Hongyan Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Ting Luo
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Zeyuan Deng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Jing Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Liufeng Zheng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Bing Zhang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| |
Collapse
|
46
|
Freiría-Martínez L, Iglesias-Martínez-Almeida M, Rodríguez-Jamardo C, Rivera-Baltanás T, Comís-Tuche M, Rodrígues-Amorím D, Fernández-Palleiro P, Blanco-Formoso M, Álvarez-Chaver P, Diz-Chaves Y, Gonzalez-Freiria N, Martín-Forero-Maestre M, Fernández-Feijoo CD, Suárez-Albo M, Fernández-Lorenzo JR, Guisán AC, Olivares JM, Spuch C. Proteomic analysis of exosomes derived from human mature milk and colostrum of mothers with term, late preterm, or very preterm delivery. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:4905-4917. [PMID: 37718950 DOI: 10.1039/d3ay01114c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
The growth and development of the human brain is a long and complex process that requires a precise sequence of genetic and molecular events. This begins in the third week of gestation with the differentiation of neural progenitor cells and extends at least until late adolescence, possibly for life. One of the defects of this development is that we know very little about the signals that modulate this sequence of events. The first 3 years of life, during breastfeeding, is one of the critical periods in brain development. In these first years of life, it is believed that neurodevelopmental problems may be the molecular causes of mental disorders. Therefore, we herein propose a new hypothesis, according to which the chemical signals that could modulate this entire complex sequence of events appear in this early period, and the molecular level study of human breast milk and colostrum of mothers who give birth to children in different gestation periods could give us information on proteins influencing this process. In this work, we collected milk and colostrum samples (term, late preterm and moderate/very preterm) and exosomes were isolated. The samples of exosomes and complete milk from each fraction were analyzed by LC-ESI-MS/MS. In this work, we describe proteins in the different fractions of mature milk and colostrum of mothers with term, late preterm, or very preterm delivery, which could be involved in the regulation of the nervous system by their functions. We describe how they differ in different types of milk, paving the way for the investigation of possible new neuroregulatory pathways as possible candidates to modulate the nervous system.
Collapse
Affiliation(s)
- Luis Freiría-Martínez
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, 36312, Spain.
- University of Vigo, Vigo, 36310, Spain
| | - Marta Iglesias-Martínez-Almeida
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, 36312, Spain.
- University of Vigo, Vigo, 36310, Spain
| | - Cynthia Rodríguez-Jamardo
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, 36312, Spain.
- University of Vigo, Vigo, 36310, Spain
| | - Tania Rivera-Baltanás
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, 36312, Spain.
- CIBERSAM, Madrid, 28029, Spain.
| | - María Comís-Tuche
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, 36312, Spain.
| | - Daniela Rodrígues-Amorím
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, 36312, Spain.
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Patricia Fernández-Palleiro
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, 36312, Spain.
| | - María Blanco-Formoso
- Department of Physical Chemistry, Singular Center for Biomedical Research (CINBIO), Universidade de Vigo, Vigo, 36310, Spain
| | - Paula Álvarez-Chaver
- Structural Determination, Proteomic and Genomic Service, CACTI, University of Vigo, Vigo, Spain
| | - Yolanda Diz-Chaves
- Laboratory of Endocrinology, Singular Center for Biomedical Research (CINBIO), Universidade de Vigo, 36310 Vigo, Spain
| | | | | | | | - María Suárez-Albo
- Neonatal Intensive Care Unit, Alvaro Cunqueiro Hospital, Vigo, 36312, Spain
| | | | | | - Jose Manuel Olivares
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, 36312, Spain.
- CIBERSAM, Madrid, 28029, Spain.
| | - Carlos Spuch
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, 36312, Spain.
- CIBERSAM, Madrid, 28029, Spain.
| |
Collapse
|
47
|
Sessitsch A, Wakelin S, Schloter M, Maguin E, Cernava T, Champomier-Verges MC, Charles TC, Cotter PD, Ferrocino I, Kriaa A, Lebre P, Cowan D, Lange L, Kiran S, Markiewicz L, Meisner A, Olivares M, Sarand I, Schelkle B, Selvin J, Smidt H, van Overbeek L, Berg G, Cocolin L, Sanz Y, Fernandes WL, Liu SJ, Ryan M, Singh B, Kostic T. Microbiome Interconnectedness throughout Environments with Major Consequences for Healthy People and a Healthy Planet. Microbiol Mol Biol Rev 2023; 87:e0021222. [PMID: 37367231 PMCID: PMC10521359 DOI: 10.1128/mmbr.00212-22] [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] [Indexed: 06/28/2023] Open
Abstract
Microbiomes have highly important roles for ecosystem functioning and carry out key functions that support planetary health, including nutrient cycling, climate regulation, and water filtration. Microbiomes are also intimately associated with complex multicellular organisms such as humans, other animals, plants, and insects and perform crucial roles for the health of their hosts. Although we are starting to understand that microbiomes in different systems are interconnected, there is still a poor understanding of microbiome transfer and connectivity. In this review we show how microbiomes are connected within and transferred between different habitats and discuss the functional consequences of these connections. Microbiome transfer occurs between and within abiotic (e.g., air, soil, and water) and biotic environments, and can either be mediated through different vectors (e.g., insects or food) or direct interactions. Such transfer processes may also include the transmission of pathogens or antibiotic resistance genes. However, here, we highlight the fact that microbiome transmission can have positive effects on planetary and human health, where transmitted microorganisms potentially providing novel functions may be important for the adaptation of ecosystems.
Collapse
Affiliation(s)
| | | | | | - Emmanuelle Maguin
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Tomislav Cernava
- University of Southampton, Faculty of Environmental and Life Sciences, Southampton, United Kingdom
| | | | | | - Paul D. Cotter
- Teagasc Food Research Centre, Moorepark, APC Microbiome Ireland and VistaMilk, Cork, Ireland
| | | | - Aicha Kriaa
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Pedro Lebre
- University of Pretoria, Pretoria, South Africa
| | - Don Cowan
- University of Pretoria, Pretoria, South Africa
| | - Lene Lange
- LL-BioEconomy, Valby, Copenhagen, Denmark
| | | | - Lidia Markiewicz
- Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Department of Immunology and Food Microbiology, Olsztyn, Poland
| | - Annelein Meisner
- Wageningen University and Research, Wageningen Research, Wageningen, The Netherlands
| | - Marta Olivares
- Institute of Agrochemistry and Food Technology, Excellence Center Severo Ochoa – Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Inga Sarand
- Tallinn University of Technology, Department of Chemistry and Biotechnology, Tallinn, Estonia
| | | | | | - Hauke Smidt
- Wageningen University and Research, Laboratory of Microbiology, Wageningen, The Netherlands
| | - Leo van Overbeek
- Wageningen University and Research, Wageningen Research, Wageningen, The Netherlands
| | | | | | - Yolanda Sanz
- Institute of Agrochemistry and Food Technology, Excellence Center Severo Ochoa – Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | | | - S. J. Liu
- Chinese Academy of Sciences, Institute of Microbiology, Beijing, China
| | - Matthew Ryan
- Genetic Resources Collection, CABI, Egham, United Kingdom
| | - Brajesh Singh
- Hawkesbury Institute for the Environment, Western Sydney University, Penrith, New South Wales, Australia
| | - Tanja Kostic
- AIT Austrian Institute of Technology GmbH, Tulln, Austria
| |
Collapse
|
48
|
Arishi RA, Lai CT, Geddes DT, Stinson LF. Impact of breastfeeding and other early-life factors on the development of the oral microbiome. Front Microbiol 2023; 14:1236601. [PMID: 37744908 PMCID: PMC10513450 DOI: 10.3389/fmicb.2023.1236601] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/25/2023] [Indexed: 09/26/2023] Open
Abstract
The oral cavity is home to the second most diverse microbiome in the human body. This community contributes to both oral and systemic health. Acquisition and development of the oral microbiome is a dynamic process that occurs over early life; however, data regarding longitudinal assembly of the infant oral microbiome is scarce. While numerous factors have been associated with the composition of the infant oral microbiome, early feeding practices (breastfeeding and the introduction of solids) appear to be the strongest determinants of the infant oral microbiome. In the present review, we draw together data on the maternal, infant, and environmental factors linked to the composition of the infant oral microbiome, with a focus on early nutrition. Given evidence that breastfeeding powerfully shapes the infant oral microbiome, the review explores potential mechanisms through which human milk components, including microbes, metabolites, oligosaccharides, and antimicrobial proteins, may interact with and shape the infant oral microbiome. Infancy is a unique period for the oral microbiome. By enhancing our understanding of oral microbiome assembly in early life, we may better support both oral and systemic health throughout the lifespan.
Collapse
Affiliation(s)
- Roaa A. Arishi
- School of Molecular Sciences, The University of Western Australia, Perth, WA, Australia
- Ministry of Health, Riyadh, Saudi Arabia
| | - Ching T. Lai
- School of Molecular Sciences, The University of Western Australia, Perth, WA, Australia
| | - Donna T. Geddes
- School of Molecular Sciences, The University of Western Australia, Perth, WA, Australia
| | - Lisa F. Stinson
- School of Molecular Sciences, The University of Western Australia, Perth, WA, Australia
| |
Collapse
|
49
|
Johnson-Hence CB, Gopalakrishna KP, Bodkin D, Coffey KE, Burr AH, Rahman S, Rai AT, Abbott DA, Sosa YA, Tometich JT, Das J, Hand TW. Stability and heterogeneity in the antimicrobiota reactivity of human milk-derived immunoglobulin A. J Exp Med 2023; 220:e20220839. [PMID: 37462916 PMCID: PMC10354535 DOI: 10.1084/jem.20220839] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 04/11/2023] [Accepted: 06/15/2023] [Indexed: 07/21/2023] Open
Abstract
Immunoglobulin A (IgA) is secreted into breast milk and is critical for both protecting against enteric pathogens and shaping the infant intestinal microbiota. The efficacy of breast milk-derived maternal IgA (BrmIgA) is dependent upon its specificity; however, heterogeneity in BrmIgA binding ability to the infant microbiota is not known. Using a flow cytometric array, we analyzed the reactivity of BrmIgA against bacteria common to the infant microbiota and discovered substantial heterogeneity between all donors, independent of preterm or term delivery. Surprisingly, we also observed intradonor variability in the BrmIgA response to closely related bacterial isolates. Conversely, longitudinal analysis showed that the antibacterial BrmIgA reactivity was relatively stable through time, even between sequential infants, indicating that mammary gland IgA responses are durable. Together, our study demonstrates that the antibacterial BrmIgA reactivity displays interindividual heterogeneity but intraindividual stability. These findings have important implications for how breast milk shapes the development of the preterm infant microbiota and protects against necrotizing enterocolitis.
Collapse
Affiliation(s)
- Chelseá B. Johnson-Hence
- Pediatrics Department, Infectious Disease Section, R.K. Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kathyayini P. Gopalakrishna
- Pediatrics Department, Infectious Disease Section, R.K. Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Darren Bodkin
- Pediatrics Department, Infectious Disease Section, R.K. Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kara E. Coffey
- Pediatrics Department, Infectious Disease Section, R.K. Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pediatrics, Division of Allergy and Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ansen H.P. Burr
- Pediatrics Department, Infectious Disease Section, R.K. Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Syed Rahman
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Systems Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ali T. Rai
- Pediatrics Department, Infectious Disease Section, R.K. Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Darryl A. Abbott
- Pediatrics Department, Infectious Disease Section, R.K. Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yelissa A. Sosa
- Pediatrics Department, Infectious Disease Section, R.K. Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Justin T. Tometich
- Pediatrics Department, Infectious Disease Section, R.K. Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jishnu Das
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Systems Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Timothy W. Hand
- Pediatrics Department, Infectious Disease Section, R.K. Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
50
|
Chen JF, Ou-Yang MC, Hsia KC, Li CM, Yeh YT, Ho HH. A Three-Arm, Randomized, Double-Blind, Placebo-Controlled Study to Evaluate the Safety of Lactobacillus salivarius AP-32 and Bifidobacterium animalis CP-9 Used Individually in Healthy Infants. Nutrients 2023; 15:3426. [PMID: 37571365 PMCID: PMC10421338 DOI: 10.3390/nu15153426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Probiotics are considered safe and beneficial to human health. However, the safety of Lactobacillus salivarius AP-32 and Bifidobacterium animalis CP-9 in infants has not been confirmed. This study was to assess the safety of long-term oral administration of L. salivarius AP-32 and B. animalis CP-9 in healthy infants compared with placebo. A three-arm, randomized, double-blind, placebo-controlled trial was conducted in healthy, full-term infants. Eighty-eight infants between 7 days and 2 months (60 ± 7 days) of age were selected and randomized to treatment with L. salivarius AP-32, B. animalis CP-9 or placebo for 4 months. The unblinding indicated subjects were randomized to receive B. animalis CP-9 (N = 28), L. salivarius AP-32 (N = 29), or placebo (N = 31). A total of 76 infants completed the 4-month treatment with fully compliance. The primary outcome was weight gain, with no significant difference in infant weight at 4 months when comparing AP-32 or CP-9 group with the placebo group, either. The head circumference and recumbent length of the CP-9 group were not significantly different from those of the placebo group. The recumbent length of the AP-32 group was slightly lower than that in the placebo group at month 4, but there was no difference between the two groups in head circumference. Overall, the growth trend of all treatments was similar without significant difference. Furthermore, there were no apparent differences between each group in digestive tolerance, the occurrence of adverse events, crying/fussing time and episodes, alpha diversity, and beta diversity. The CP-9 group showed a significant increase in the abundance of the Bacteroides genus, while the AP-32 group demonstrated a significant increase in the abundance of the Lactobacillus genus when comparing the two probiotic groups. Our study findings indicate that the oral administration of both AP-32 and CP-9 strains has a positive impact on the maintenance of a healthy gut flora in infants. Long-term use of L. salivarius AP-32 or B. animalis CP-9 is safe for infants from 7 days to 6 months of age.
Collapse
Affiliation(s)
- Jui-Fen Chen
- Department of Research and Design, Glac Biotech Co., Ltd., Tainan 744, Taiwan; (J.-F.C.); (K.-C.H.); (C.-M.L.)
| | - Mei-Chen Ou-Yang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan;
| | - Ko-Chiang Hsia
- Department of Research and Design, Glac Biotech Co., Ltd., Tainan 744, Taiwan; (J.-F.C.); (K.-C.H.); (C.-M.L.)
| | | | - Ching-Min Li
- Department of Research and Design, Glac Biotech Co., Ltd., Tainan 744, Taiwan; (J.-F.C.); (K.-C.H.); (C.-M.L.)
| | - Yao-Tsung Yeh
- Aging and Disease Prevention Research Center, Fooyin University, Kaohsiung 831, Taiwan;
- Department of Medical Laboratory Science and Biotechnology, Fooyin University, Kaohsiung 831, Taiwan
| | - Hsieh-Hsun Ho
- Department of Research and Design, Glac Biotech Co., Ltd., Tainan 744, Taiwan; (J.-F.C.); (K.-C.H.); (C.-M.L.)
| |
Collapse
|