1
|
Al-Beltagi M. Human milk oligosaccharide secretion dynamics during breastfeeding and its antimicrobial role: A systematic review. World J Clin Pediatr 2025; 14. [DOI: 10.5409/wjcp.v14.i2.104797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 02/19/2025] [Accepted: 02/27/2025] [Indexed: 03/18/2025] Open
Abstract
BACKGROUND
Human milk oligosaccharides (HMOs) are bioactive components of breast milk with diverse health benefits, including shaping the gut microbiota, modulating the immune system, and protecting against infections. HMOs exhibit dynamic secretion patterns during lactation, influenced by maternal genetics and environmental factors. Their direct and indirect antimicrobial properties have garnered significant research interest. However, a comprehensive understanding of the secretion dynamics of HMOs and their correlation with antimicrobial efficacy remains underexplored.
AIM
To synthesize current evidence on the secretion dynamics of HMOs during lactation and evaluate their antimicrobial roles against bacterial, viral, and protozoal pathogens.
METHODS
A systematic search of PubMed, Scopus, Web of Science, and Cochrane Library focused on studies investigating natural and synthetic HMOs, their secretion dynamics, and antimicrobial properties. Studies involving human, animal, and in vitro models were included. Data on HMO composition, temporal secretion patterns, and mechanisms of antimicrobial action were extracted. Quality assessment was performed using validated tools appropriate for study design.
RESULTS
A total of 44 studies were included, encompassing human, animal, and in vitro research. HMOs exhibited dynamic secretion patterns, with 2′-fucosyllactose (2′-FL) and lacto-N-tetraose peaking in early lactation and declining over time, while 3-fucosyllactose (3-FL) increased during later stages. HMOs demonstrated significant antimicrobial properties through pathogen adhesion inhibition, biofilm disruption, and enzymatic activity impairment. Synthetic HMOs, including bioengineered 2′-FL and 3-FL, were structurally and functionally comparable to natural HMOs, effectively inhibiting pathogens such as Pseudomonas aeruginosa , Escherichia coli , and Campylobacter jejuni . Additionally, HMOs exhibited synergistic effects with antibiotics, enhancing their efficacy against resistant pathogens.
CONCLUSION
HMOs are vital in antimicrobial defense, supporting infant health by targeting various pathogens. Both natural and synthetic HMOs hold significant potential for therapeutic applications, particularly in infant nutrition and as adjuncts to antibiotics. Further research, including clinical trials, is essential to address gaps in knowledge, validate findings, and explore the broader applicability of HMOs in improving maternal and neonatal health.
Collapse
Affiliation(s)
- Mohammed Al-Beltagi
- Department of Paediatrics, Faculty of Medicine, Tanta University, Tanta 31511, Alghrabia, Egypt
- Department of Pediatric, University Medical Center, King Abdulla Medical City, Arabian Gulf University, Manama 26671, Bahrain
| |
Collapse
|
2
|
Wang X, Tong J, Li H, Lu M, Liu Y, Gan H, Wang Y, Geng M, Qie X, Wu X, Gao H, Zhu B, Tao S, Tao X, Yan S, Gao G, Wu X, Huang K, Cao Y, Tao F. Sex-and stage-specific effect of prenatal exposure to organophosphate esters with children's physical growth patterns and adiposity rebound timing: Modification by breastfeeding. JOURNAL OF HAZARDOUS MATERIALS 2025; 493:138309. [PMID: 40252324 DOI: 10.1016/j.jhazmat.2025.138309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 04/14/2025] [Accepted: 04/14/2025] [Indexed: 04/21/2025]
Abstract
INTRODUCTION Exploring the stage-specific effects of prenatal exposure to organophosphate esters (OPEs) on offspring growth and developmental trajectories is critical for early-life health management. METHODS Based on 2519 mother-child dyads from the Ma'anshan Birth Cohort, we examined the concentrations of OPEs in maternal urine during the three trimesters. Seventeen follow-up visits were made to the children, and physical data were collected. A grouped trajectory model was used to fit the growth trajectories. RESULTS First-trimester bis(2-butoxyethyl) phosphate (BBOEP) was inversely associated with the children's adiposity rebound (AR) timing (β = -0.33, 95 % CI: -0.65, -0.01), and the ORs (95 % CIs) for early age at AR for each doubling of BBOEP and dibutyl phosphate (DBP) were 1.07 (1.00, 1.14) and 1.12 (1.03, 1.22), respectively. BBOEP increased the risk of a high-stable BMI-for-age z score (BMIz) group (OR = 1.18, 95 % CI: 1.01, 1.39), whereas tris(2-chloroethyl) phosphate (TCEP) and bis(2-ethylhexyl) phosphate reduced this risk. Diphenyl phosphate (OR = 0.74, 95 % CI: 0.59, 0.94) and aromatic OPEs (OR = 0.70, 95 % CI: 0.54, 0.90) reduced the odds of an extreme-high body fat group. TCEP also reduced the risk of a high body fat percentage group (p < 0.05). There appeared to be sex and ester bond differences in these associations, and breastfeeding could counteract the association between the OPEs and growth trajectories. No mixed effects of OPEs on BMIz trajectories were found. CONCLUSIONS The present study identified a heterogeneous association between OPE exposure during pregnancy and AR timing and physical growth patterns in offspring. Future studies are needed involving more regions and populations, with consideration of other developmentally toxic compounds, to obtain more reliable and comprehensive results.
Collapse
Affiliation(s)
- Xing Wang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China
| | - Juan Tong
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China; MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei, Anhui 230032, China
| | - Han Li
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China
| | - Mengjuan Lu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China; MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei, Anhui 230032, China
| | - Yuan Liu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China
| | - Hong Gan
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China; MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei, Anhui 230032, China
| | - Yifan Wang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China
| | - Menglong Geng
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China; MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei, Anhui 230032, China
| | - Xuejiao Qie
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China; MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei, Anhui 230032, China
| | - Xiulong Wu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China; MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, Anhui 230032, China; Ma'anshan Maternal and Child Health Care Hospital, Ma'anshan, Anhui 243011, China
| | - Hui Gao
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei, Anhui 230032, China; Department of Pediatrics, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Beibei Zhu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China; MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei, Anhui 230032, China
| | - Shuman Tao
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei, Anhui 230032, China
| | - Xingyong Tao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China; MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei, Anhui 230032, China
| | - Shuangqin Yan
- Ma'anshan Maternal and Child Health Care Hospital, Ma'anshan, Anhui 243011, China
| | - Guopeng Gao
- Ma'anshan Maternal and Child Health Care Hospital, Ma'anshan, Anhui 243011, China
| | - Xiaoyan Wu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China; MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei, Anhui 230032, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, Anhui 230032, China
| | - Kun Huang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China; MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei, Anhui 230032, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, Anhui 230032, China
| | - Yunxia Cao
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, Anhui 230032, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, Anhui 230032, China
| | - Fangbiao Tao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China; MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei, Anhui 230032, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, Anhui 230032, China.
| |
Collapse
|
3
|
Zhu L, Zhang Z, Luo T, Li H, Deng Z, Li J, Zheng L, Liao J, Wang M, Zhang B. Cognitive and behavioral benefits of 2'-fucosyllactose in growing mice: the roles of 5-hydroxytryptophan and gut microbiota. MICROBIOME 2025; 13:97. [PMID: 40221776 PMCID: PMC11992797 DOI: 10.1186/s40168-025-02094-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 03/17/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND 2'-Fucosyllactose (2'-FL) is one of the major oligosaccharides found in human breast milk, with several recognized beneficial effects on the host. Extensive research has indicated positive effects of 2'-FL on cognitive development in the brain, yet its molecular mechanisms have remained elusive. This study aimed to assess the impact of 2'-FL on the gut-brain axis microbiota and cognitive function in growing mice, along with its potential mechanisms of action. RESULTS Following long-term supplementation for 4 weeks, 2'-FL was found to enhance cognitive memory function in growing mice (3 weeks old) as assessed through Y-maze, novel object recognition, and water maze tests. Analysis via 16S rRNA sequencing revealed significant alterations in gut microbiota diversity and composition induced by 2'-FL, notably increasing the relative abundance of Bacteroides and Lactobacillus genera. Additionally, 2'-FL significantly elevated levels of 5-hydroxytryptamine (5-HT) and 5-hydroxytryptophan (5-HTP) in the hippocampal tissue. However, antibiotic intervention abolished the cognitive advantage conferred by 2'-FL, highlighting the critical role of gut microbiota in mediating its effects. Similarly, short-term supplementation with 2'-FL for 7 days indicated rapid changes in gut microbiota composition preceding cognitive improvements, further suggesting a potential causal relationship between gut microbiota characteristics and cognition. Further, in vitro experiments with mouse feces suggested that 2'-FL may influence tryptophan hydroxylase levels in the gut microbiota and inhibit the activity of 5-hydroxytryptophan decarboxylase, potentially leading to increased accumulation of 5-HTP. Additionally, 2'-FL may indirectly impact tryptophan hydroxylase levels in enterochromaffin cells by promoting short-chain fatty acid production, which could support 5-HTP synthesis. Elevated 5-HTP produced by the gut system enters the bloodstream, crosses the blood-brain barrier, and may potentially enhance brain 5-HT levels. CONCLUSION This study offers preliminary evidence that the cognitive-promoting effects of 2'-FL in mice may be closely associated with gut microbiota and 5-HT. The findings suggest that 2'-FL contribute to cognitive development in growing mice, potentially by modulating gut microbiota and enhancing 5-HT levels in the brain. Video Abstract.
Collapse
Affiliation(s)
- Liuying Zhu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China
| | - Zhiyi Zhang
- 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
- International Institute of Food Innovation, Nanchang University, Nanchang , Jiangxi, 330051, China
| | - Hongyan Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China
- International Institute of Food Innovation, Nanchang University, Nanchang , Jiangxi, 330051, China
| | - Zeyuan Deng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China.
- International Institute of Food Innovation, Nanchang University, Nanchang , Jiangxi, 330051, China.
| | - Jing Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China
- International Institute of Food Innovation, Nanchang University, Nanchang , Jiangxi, 330051, China
| | - Liufeng Zheng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China
- International Institute of Food Innovation, Nanchang University, Nanchang , Jiangxi, 330051, China
| | - Jinqiang Liao
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China
| | - Minghui Wang
- 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.
- International Institute of Food Innovation, Nanchang University, Nanchang , Jiangxi, 330051, China.
| |
Collapse
|
4
|
Catić Đorđević A, Stefanović A, Kovačević M, Vezmar Kovačević S, Stanković L, Miljković B. Community pharmacists' support for nursing mothers in Serbia: potential cost savings due to breastfeeding continuation. BMC Health Serv Res 2025; 25:523. [PMID: 40205538 PMCID: PMC11983895 DOI: 10.1186/s12913-025-12523-0] [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: 11/14/2024] [Accepted: 03/05/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Despite many health benefits to infants, the overall breastfeeding rate remains low among Serbian women. The community pharmacists' efforts aimed at supporting nursing mothers are important. The aim of the study was an evaluation of the pharmacists' training for breastfeeding support and their perception about its usefulness. The second aim was an evaluation of the provided structured pharmaceutical care service (SPS) over four months, through a calculation of theoretical cost savings for counseled families in case of the risk for introduction of commercial milk formula (CMF). METHODS The continuation of breastfeeding instead of starting with CMF was theoretically monetized through cost savings analysis, which is based on energy equivalent human milk and CMF, regardless of the differences in health, social and ecology outcomes of breastfeeding. Cost savings for the counseled families were calculated as financial differences between continuing breastfeeding and switching to CMF. RESULTS The 256 pharmacists showed high satisfaction rates with the quality and the usefulness of the training (mean scores: content value 4.89 ± 0.51, technical value 4.91 ± 0.41, program value 4.96 ± 0.20, and usefulness value 4.94 ± 0.25, respectively), as well as a statistically significant higher level of knowledge (p < 0.001 for each of the 10 questions). The prospective observational study enrolled 256 community pharmacists who voluntarily underwent training aimed to breastfeeding support. Of all the trained pharmacists, 151 (59%) actively provided 1,243 SPS focused on breastfeeding support. Of all, 599 mothers of children under 6 months of age received SPS, while 590 provided full data In 275 cases (22.1%), a risk for early breastfeeding cessation was identified. The average age of children whose mothers had undergone SPS was 2.83 months, and the total average potential cost savings until 6 months of children age was 35,884.80 RSD (approximately 306 EUR) for average of 3.17 months. CONCLUSIONS The training of pharmacists adds significant value to their knowledge and skills for breastfeeding support, which may lead to potential cost savings for families.
Collapse
|
5
|
Abate BB, Tusa BS, Sendekie AK, Araya FG, Bizuayehu MA, Walle GT, Kitaw TA, Tilahun BD, Alamaw AW, Zemariam AB, Kassaw A, Kassie AM, Yilak G, Bizuneh FK, Dachew B. Non-exclusive breastfeeding is associated with pneumonia and asthma in under-five children: an umbrella review of systematic review and meta-analysis. Int Breastfeed J 2025; 20:18. [PMID: 40134007 PMCID: PMC11938590 DOI: 10.1186/s13006-025-00712-w] [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: 05/03/2024] [Accepted: 03/16/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Despite numerous reviews examining the impact of exclusive breastfeeding on preventing childhood pneumonia and asthma, a comprehensive and up-to-date synthesis is lacking. This umbrella review aims to consolidate the current evidence on the link between non-exclusive breastfeeding and the risk of pneumonia and asthma in under-five children. METHODS A comprehensive search was conducted in PubMed, Embase, Scopus, Web of Science, the Cochrane Database of Systematic Reviews, and Google Scholar to identify systematic review and meta-analysis (SRM) studies evaluating the effect of exclusive breastfeeding on preventing childhood pneumonia and asthma globally. The latest search was conducted on January 25/2025. The quality of the included studies was assessed using the Assessment of Multiple Systematic Reviews Two (AMSTAR-2) tool. A weighted inverse variance random-effects model was employed to generate pooled estimates. Summary effect estimates were expressed using odds ratios (OR) with 95% confidence intervals (CI). We evaluated the quality of evidence for each association using the Grading of Recommendations, Assessment, Development, and Evaluations (GRADE) framework, categorising it as convincing (class I), highly suggestive (class II), suggestive (class III), and weak (class IV). RESULTS Twelve SRMs, including 270 primary studies with over ten million participants, were analysed. The random-effects model revealed a highly suggestive association between non-exclusive breastfeeding and an increase in the risk of pneumonia (OR 2.34; 95% CI 1.89, 2.78, GRADE: highly suggestive). Similarly, there was highly suggestive evidence that non-exclusive breastfeeding was associated with a 29% higher risk of childhood asthma (OR 1.21; 95% CI 1.07, 1.34, GRADE: highly suggestive). CONCLUSION Our results highlighted that non-exclusive breastfeeding is associated with an increased risk of pneumonia and asthma in under-five children. These findings emphasise the critical role of exclusive breastfeeding in reducing the risk of respiratory health issues, highlighting the need for policies and initiatives that promote breastfeeding as a key strategy for improving children's health outcomes.
Collapse
Affiliation(s)
- Biruk Beletew Abate
- School of Population Health, Curtin University, Perth, WA, Australia.
- College of Medicine and Health Sciences, Woldia University, Woldia, Ethiopia.
| | - Biruk Shalmeno Tusa
- School of Population Health, Curtin University, Perth, WA, Australia
- Department of Epidemiology and Biostatistics, College of Health and Medical Sciences, Haramaya University, Haramaya, Ethiopia
| | - Ashenafi Kibret Sendekie
- Department of Clinical Pharmacy, School of Pharmacy, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
- School of Pharmacy, Curtin Medical School, Faculty of Health Sciences, Curtin University, Bentley, WA, Australia
| | | | | | - Getachew Tesfaw Walle
- Department of Nursing, College of Health Science, Woldia University, Woldia, Ethiopia
| | - Tegene Atamenta Kitaw
- Department of Nursing, College of Health Science, Woldia University, Woldia, Ethiopia
| | - Befkad Derese Tilahun
- Department of Nursing, College of Health Science, Woldia University, Woldia, Ethiopia
| | | | - Alemu Birara Zemariam
- Department of Nursing, College of Health Science, Woldia University, Woldia, Ethiopia
| | - Amare Kassaw
- Department of Nursing, College of Health Science, Debre Tabor University, Debra Tabor, Ethiopia
| | | | - Gizachew Yilak
- Department of Nursing, College of Health Science, Woldia University, Woldia, Ethiopia
| | | | - Berihun Dachew
- School of Population Health, Curtin University, Perth, WA, Australia
- enAble Institute, Curtin University, Perth, WA, Australia
| |
Collapse
|
6
|
Mikles B, Schmidt CJ, Benbow ME, Jordan HR, Pechal JL. Potential postmortem microbial biomarkers of infant and younger children death investigation. J Forensic Sci 2025; 70:607-618. [PMID: 39682072 PMCID: PMC11874197 DOI: 10.1111/1556-4029.15677] [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: 06/03/2024] [Revised: 08/08/2024] [Accepted: 11/13/2024] [Indexed: 12/18/2024]
Abstract
Microbial communities associated with the human body are highly dynamic and reflect the host environment and lifestyle over time. Studies show death is no exception, with data demonstrating similar antemortem and postmortem microbiomes up to 48 h following death. These predictable microbial biomarkers can inform death investigation by helping to estimate the postmortem interval and build models to identify cause and manner of death. However, no attempts have been made to model potential microbial biomarkers in pediatric (≤2 years) deaths. This study provided a cross-sectional survey of the microbiota of 53 pediatric cases (black, white, both sexes) seen in Wayne County, Michigan. Autopsy cases represented accidents, homicides, or natural causes. Postmortem microbiome were collected by swabbing the eyes, ears, nose, mouth, umbilicus, brain, rectum, trabecular space, and cardiac blood. 16S rRNA sequence analyses indicated that sex, race, age, body site, and manner of death (MOD) had significant effects on microbiome composition, with significant interactions among MOD, race, and age. Amplicon sequence variants identified intra- and interhost dispersion of the postmortem microbiome depending on death circumstance. Among manners of death, non-accidental deaths were significantly distinct from all other deaths, and among body sites the rectum was distinct in its microbial composition. There is a real need for robust postmortem microbiome before it can be standardized as a practical tool for use in forensic investigation or public health. These results inform postmortem microbial variability during pediatric death investigation that contributes to a larger effort to understand the postmortem microbiome.
Collapse
Affiliation(s)
- Bethany Mikles
- Department of EntomologyMichigan State UniversityEast LansingMichiganUSA
| | - Carl J. Schmidt
- Department of PathologyUniversity of MichiganAnn ArborMichiganUSA
| | - M. Eric Benbow
- Department of EntomologyMichigan State UniversityEast LansingMichiganUSA
- Department of Osteopathic Medical SpecialtiesMichigan State UniversityEast LansingMichiganUSA
- AgBioResearchMichigan State UniversityEast LansingMichiganUSA
- Ecology, Evolution and Behavior ProgramMichigan State UniversityEast LansingMichiganUSA
| | - Heather R. Jordan
- Department of Biological SciencesMississippi State UniversityStarkvilleMississippiUSA
| | - Jennifer L. Pechal
- Department of EntomologyMichigan State UniversityEast LansingMichiganUSA
| |
Collapse
|
7
|
Dos Santos IBL, Fioretto MN, Jorge MS, Barata LA, Ribeiro IT, Franzolin AML, Stoppa EG, Mattos R, Portela LMF, Emílio Silva MT, Dos Santos SAA, de Arruda Miranda JR, Hiruma Lima CA, Justulin LA. Maternal protein restriction impairs intestinal morphophysiology and antioxidant system in young male offspring rats. Exp Cell Res 2025; 446:114464. [PMID: 39986598 DOI: 10.1016/j.yexcr.2025.114464] [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: 12/21/2024] [Revised: 02/10/2025] [Accepted: 02/16/2025] [Indexed: 02/24/2025]
Abstract
The developmental origins of health and disease (DOHaD) concept suggests that adverse conditions during gestation can influence the development and function of multiple organs, including the gastrointestinal tract. Maternal protein restriction (MPR) exposure has been associated with negative effects on reproduction, the endocrine system, and liver metabolic health. However, limited research has explored the impact of MPR on the offspring's intestinal morphophysiology. This study investigated the effects of gestational and lactational MPR on the duodenum and colon of young male offspring rats at postnatal (PND)21. We hypothesize that MPR affects intestinal morphophysiology and development early in life. Our findings revealed tachygastria in offspring exposed to MPR. The ultrastructural analysis uncovered a reduction in goblet cell numbers and changes in collagen deposition in the duodenum and colon. We also identified imbalances in inflammatory markers (IL-6 and TGF-β1) and antioxidant enzymes (CAT and SOD). These results demonstrate that MPR significantly affects gastrointestinal morphophysiology early in life by disrupting gastric motility and altering duodenal and colonic histoarchitecture, antioxidant defense, and inflammatory pathways. Such alterations may predispose the descendants to long-term gastrointestinal disorders, underscoring the importance of further research on the developmental origins of intestinal health and disease.
Collapse
Affiliation(s)
| | - Matheus Naia Fioretto
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Miguel Silingardi Jorge
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Luísa Annibal Barata
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Isabelle Tenori Ribeiro
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | | | - Erick Guilherme Stoppa
- Department of Biophysics and Pharmacology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Renato Mattos
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Luiz Marcos Frediane Portela
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Maycon Tavares Emílio Silva
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Sérgio Alexandre Alcântara Dos Santos
- Department of Biophysics and Pharmacology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil; Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | | | - Clélia Akiko Hiruma Lima
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Luis Antonio Justulin
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil.
| |
Collapse
|
8
|
Eisen A, Kiernan MC. The Neonatal Microbiome: Implications for Amyotrophic Lateral Sclerosis and Other Neurodegenerations. Brain Sci 2025; 15:195. [PMID: 40002527 PMCID: PMC11852589 DOI: 10.3390/brainsci15020195] [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: 01/13/2025] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Most brain development occurs in the "first 1000 days", a critical period from conception to a child's second birthday. Critical brain processes that occur during this time include synaptogenesis, myelination, neural pruning, and the formation of functioning neuronal circuits. Perturbations during the first 1000 days likely contribute to later-life neurodegenerative disease, including sporadic amyotrophic lateral sclerosis (ALS). Neurodevelopment is determined by many events, including the maturation and colonization of the infant microbiome and its metabolites, specifically neurotransmitters, immune modulators, vitamins, and short-chain fatty acids. Successful microbiome maturation and gut-brain axis function depend on maternal factors (stress and exposure to toxins during pregnancy), mode of delivery, quality of the postnatal environment, diet after weaning from breast milk, and nutritional deficiencies. While the neonatal microbiome is highly plastic, it remains prone to dysbiosis which, once established, may persist into adulthood, thereby inducing the development of chronic inflammation and abnormal excitatory/inhibitory balance, resulting in neural excitation. Both are recognized as key pathophysiological processes in the development of ALS.
Collapse
Affiliation(s)
- Andrew Eisen
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Matthew C. Kiernan
- Neuroscience Research Australia, University of New South Wales, Randwick, Sydney, NSW 2031, Australia;
| |
Collapse
|
9
|
Capra ME, Aliverti V, Bellani AM, Berzieri M, Montani AG, Pisseri G, Sguerso T, Esposito S, Biasucci G. Breastfeeding and Non-Communicable Diseases: A Narrative Review. Nutrients 2025; 17:511. [PMID: 39940369 PMCID: PMC11819769 DOI: 10.3390/nu17030511] [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: 01/08/2025] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025] Open
Abstract
INTRODUCTION Breastfeeding plays a fundamental role in newborns' and infants' health. Breast milk's protective power against malnutrition and its positive effect on neurological and physical development are well established and are reflected in the policy statements of all major pediatric health entities. However, breastfeeding also plays an important role in the prevention of so-called non-communicable diseases, such as obesity, hypertension, dyslipidemia, and autoimmune diseases. METHODS This narrative review aims to analyze the effect of breastfeeding and breast milk on the development of non-communicable diseases, with a special focus on weight excess, dyslipidemia, allergy, and gastrointestinal diseases. This narrative review was carried out through three steps: executing the search, examining abstracts and full texts, and analyzing results. To achieve this, the databases PubMed, EMBASE, Scopus, ScienceDirect, Web of Science, and Google Scholar were explored to collect and select publications from 1990 to 2024 to find pertinent studies in line with this review's development. The search included randomized placebo-controlled trials, controlled clinical trials, double-blind, randomized controlled studies, and systematic reviews. A total of 104 manuscripts were ultimately included in the analysis. RESULTS Breastfeeding is associated with a decreased vulnerability to early viral infections or chronic inflammatory conditions during preschool years, a reduced incidence of weight excess, and likely lower cholesterol concentration, besides having a small protective effect against systolic blood hypertension. CONCLUSIONS Pediatricians must promote breastfeeding, support the mother-infant dyad, and consider breast milk as a real "health voucher" that can last lifelong. However, further studies are needed to better define the extent and duration of breastfeeding's protective power in this context.
Collapse
Affiliation(s)
- Maria Elena Capra
- Pediatrics and Neonatology Unit, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy; (M.E.C.); (G.B.)
| | - Valentina Aliverti
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Arianna Maria Bellani
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Martina Berzieri
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Anna Giuseppina Montani
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Gianlorenzo Pisseri
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Tullia Sguerso
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Susanna Esposito
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Giacomo Biasucci
- Pediatrics and Neonatology Unit, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy; (M.E.C.); (G.B.)
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| |
Collapse
|
10
|
Liu T, Yuan Y, Wei J, Chen J, Zhang F, Chen J, Zhang J. Association of breast milk microbiota and metabolites with neonatal jaundice. Front Pediatr 2025; 12:1500069. [PMID: 39834492 PMCID: PMC11743730 DOI: 10.3389/fped.2024.1500069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025] Open
Abstract
Background Breast milk is the primary source of nutrition during early life, and existing research indicates that the development of jaundice in breastfed newborns may be linked to specific nutrients or bioactive substances present in breast milk. However, the association between the microbiota and small-molecule metabolites in breast milk and the development of neonatal jaundice remains unproven. This study aimed to investigate the development of jaundice in breastfed neonates in relation to breast milk microbiota and metabolites. Methods Based on the conditions of exclusive breastfeeding, we selected healthy newborns without significant jaundice and their mothers on day 4 (96-120 h after birth) as the healthy control group, and jaundiced newborns and their mothers as the jaundice group. Breast milk samples were collected from mothers in both groups on postnatal day 4 and analyzed for microbiota and small-molecule metabolites using 16S rRNA gene sequencing and an liquid chromatography-tandem mass spectrometry techniques. Results A total of 104 mother-child pairs were included in the study, of which 51 pairs were in the healthy control group and the other 53 pairs were in the jaundice group. Our results demonstrated that there was no significant difference between the species composition and diversity of the breast milk flora in the healthy control and jaundice groups. At the genus level, the abundance of Lactobacillus, Ackermannia, and Bifidobacterium was significantly higher in the breast milk of the healthy control group than in the jaundice group. Metabolomics analysis revealed a total of 27 significantly different metabolites between the two groups. Notably, breast milk from the healthy control group had elevated levels of 24 metabolites, predominantly lipids family, including sphingolipids, phospholipids, and fatty acid derivatives. Conclusion This study suggests that there is a link between the development of neonatal jaundice and breast milk microbiota and metabolites. Breast milk from mothers of healthy newborns contains higher levels of beneficial bacteria and lipid family compared to mothers of newborns with jaundice. This study offers new insights into the relationship between breastfeeding and neonatal jaundice.
Collapse
Affiliation(s)
- TianYu Liu
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
- Department of Pediatrics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanhan Yuan
- Department of Pediatrics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinying Wei
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
- Department of Pediatrics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiayi Chen
- Department of Pediatrics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Zhang
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
- Department of Pediatrics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juanjuan Chen
- Department of Pediatrics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinping Zhang
- Department of Pediatrics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Park PG, Heo JS, Ahn YH, Kang HG. Association between Exclusive Breastfeeding and the Incidence of Childhood Nephrotic Syndrome. J Pediatr 2025; 276:114266. [PMID: 39218209 DOI: 10.1016/j.jpeds.2024.114266] [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: 05/30/2024] [Revised: 08/21/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE To assess the relationship between breastfeeding and the risk of developing nephrotic syndrome using a population-based nationwide birth cohort in Korea. STUDY DESIGN This nationwide cohort study utilized data from the National Health Information Database and the National Health Screening Program for Infants and Children. The study included all children born between January 1, 2010, and December 31, 2018, who underwent their first health screening, which included a specific questionnaire on breastfeeding between 4 and 6 months of age. Associations between nephrotic syndrome and exclusive breastfeeding were estimated using adjusted hazard ratios (aHR) derived from Cox proportional hazards models, adjusted for sociodemographic variables, with follow-up until the occurrence of nephrotic syndrome, 8 years postindex date, death, or December 31, 2022, whichever was first. RESULTS The study population comprised 1 787 774 children (median follow-up: 7.96 years; IQR: 6.31-8.00 years), including 612 556 exclusively breastfed and 1 175 218 formula-fed children. Exclusive breastfeeding was associated with a decreased risk of developing nephrotic syndrome (aHR: 0.80; 95% CI: 0.69-0.93). Subgroup analysis stratified by sex mirrored the overall findings, although statistical significance was not observed in girls (boys: aHR, 0.75; 95% CI, 0.62-0.92; girls: aHR, 0.87; 95% CI, 0.70-1.09). Sensitivity analysis confirmed these results. CONCLUSIONS Exclusive breastfeeding was associated with a 20% reduced risk of developing nephrotic syndrome up to 8 years of age.
Collapse
Affiliation(s)
- Peong Gang Park
- Department of Pediatrics, Ajou University School of Medicine, Suwon, South Korea; Department of Translational Medicine, Seoul National University College of Medicine, Seoul, South Korea.
| | - Ju Sun Heo
- Department of Pediatrics, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Yo Han Ahn
- Department of Pediatrics, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Hee Gyung Kang
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, South Korea; Department of Pediatrics, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
12
|
Jeyaraman N, Jeyaraman M, Dhanpal P, Ramasubramanian S, Ragavanandam L, Muthu S, Santos GS, da Fonseca LF, Lana JF. Gut microbiome and orthopaedic health: Bridging the divide between digestion and bone integrity. World J Orthop 2024; 15:1135-1145. [PMID: 39744736 PMCID: PMC11686522 DOI: 10.5312/wjo.v15.i12.1135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 10/12/2024] [Accepted: 11/13/2024] [Indexed: 12/17/2024] Open
Abstract
The gut microbiome, a complex ecosystem of microorganisms in the digestive tract, has emerged as a critical factor in human health, influencing metabolic, immune, and neurological functions. This review explores the connection between the gut microbiome and orthopedic health, examining how gut microbes impact bone density, joint integrity, and skeletal health. It highlights mechanisms linking gut dysbiosis to inflammation in conditions such as rheumatoid arthritis and osteoarthritis, suggesting microbiome modulation as a potential therapeutic strategy. Key findings include the microbiome's role in bone metabolism through hormone regulation and production of short-chain fatty acids, crucial for mineral absorption. The review also considers the effects of diet, probiotics, and fecal microbiota transplantation on gut microbiome composition and their implications for orthopedic health. While promising, challenges in translating microbiome research into clinical practice persist, necessitating further exploration and ethical consideration of microbiome-based therapies. This interdisciplinary research aims to link digestive health with musculoskeletal integrity, offering new insights into the prevention and management of bone and joint diseases.
Collapse
Affiliation(s)
- Naveen Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600077, Tamil Nadu, India
- Department of Orthopaedics, Orthopaedic Research Group, Coimbatore 641045, Tamil Nadu, India
| | - Madhan Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600077, Tamil Nadu, India
- Department of Orthopaedics, Orthopaedic Research Group, Coimbatore 641045, Tamil Nadu, India
- Department of Orthopaedics, Brazilian Institute of Regenerative Medicine, Indaiatuba 13334-170, São Paulo, Brazil
| | - Priya Dhanpal
- Department of General Medicine, Government Medical College, Omandurar Government Estate, Chennai 600002, Tamil Nadu, India
| | - Swaminathan Ramasubramanian
- Department of General Medicine, Government Medical College, Omandurar Government Estate, Chennai 600002, Tamil Nadu, India
| | - Lavanya Ragavanandam
- Department of Pharmacology, Faculty of Medicine - Sri Lalithambigai Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600095, Tamil Nadu, India
| | - Sathish Muthu
- Department of Orthopaedics, Orthopaedic Research Group, Coimbatore 641045, Tamil Nadu, India
- Department of Orthopaedics, Government Medical College and Hospital, Karur 639004, Tamil Nadu, India
- Department of Biotechnology, Karpagam Academy of Higher Education, Coimbatore 641021, Tamil Nadu, India
| | - Gabriel Silva Santos
- Department of Orthopaedics, Brazilian Institute of Regenerative Medicine, Indaiatuba 13334-170, São Paulo, Brazil
| | - Lucas Furtado da Fonseca
- Department of Orthopaedics, Brazilian Institute of Regenerative Medicine, Indaiatuba 13334-170, São Paulo, Brazil
| | - José Fábio Lana
- Department of Orthopaedics, Brazilian Institute of Regenerative Medicine, Indaiatuba 13334-170, São Paulo, Brazil
| |
Collapse
|
13
|
Kuniyoshi Y. The Lack of an Association of Breastfeeding with the Development of Childhood Intussusception: A Nationwide Birth Cohort Survey in Japan. Breastfeed Med 2024; 19:924-931. [PMID: 39324892 DOI: 10.1089/bfm.2024.0264] [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] [Indexed: 09/27/2024]
Abstract
Background: This study investigated the association between feeding practices and the development of childhood intussusception. Materials and Methods: We conducted secondary data analyses using the Longitudinal Survey of Newborns in the 21st Century in Japan. We performed multivariable logistic regression analyses to examine the association between feeding practice and intussusception development in children aged between 6 and 18 months. We used the following variables as potential confounders: gender, gestational age, birth weight, singleton or multiple births, parity, maternal age at delivery, maternal smoking status, and paternal smoking status. Furthermore, we performed multivariable logistic regression analyses to examine the association between breastfeeding duration and intussusception development. Results: In total, 31,802 children were analyzed in this study. The annual incidence of intussusception was 1.6 cases per 1,000 children aged between 6 and 18 months. No significant association was found between exclusive breastfeeding and the development of intussusception, compared with exclusive formula feeding (odds ratio, 1.64; 95% confidence interval, 0.32-30.0). Furthermore, no significant association was observed between breastfeeding duration and intussusception development. Conclusions: Our findings demonstrated no association between breastfeeding and the development of childhood intussusception.
Collapse
Affiliation(s)
- Yasutaka Kuniyoshi
- Department of Social Services and Healthcare Management, International University of Health and Welfare, Otawara, Japan
| |
Collapse
|
14
|
Catassi G, Mateo SG, Occhionero AS, Esposito C, Giorgio V, Aloi M, Gasbarrini A, Cammarota G, Ianiro G. The importance of gut microbiome in the perinatal period. Eur J Pediatr 2024; 183:5085-5101. [PMID: 39358615 PMCID: PMC11527957 DOI: 10.1007/s00431-024-05795-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024]
Abstract
This narrative review describes the settlement of the neonatal microbiome during the perinatal period and its importance on human health in the long term. Delivery methods, maternal diet, antibiotic exposure, feeding practices, and early infant contact significantly shape microbial colonization, influencing the infant's immune system, metabolism, and neurodevelopment. By summarizing two decades of research, this review highlights the microbiome's role in disease predisposition and explores interventions like maternal vaginal seeding and probiotic and prebiotic supplementation that may influence microbiome development. CONCLUSION The perinatal period is a pivotal phase for the formation and growth of the neonatal microbiome, profoundly impacting long-term health outcomes. WHAT IS KNOWN • The perinatal period is a critical phase for the development of the neonatal microbiome, with factors such as mode of delivery, maternal diet, antibiotic exposure, and feeding practices influencing its composition and diversity, which has significant implications for long-term health. • The neonatal microbiome plays a vital role in shaping the immune system, metabolism, and neurodevelopment of infants. WHAT IS NEW • Recent studies have highlighted the potential of targeted interventions, such as probiotic and prebiotic supplementation, and innovative practices like maternal vaginal seeding, to optimize microbiome development during the perinatal period. • Emerging evidence suggests that specific bacterial genera and species within the neonatal microbiome are associated with reduced risks of developing chronic conditions, indicating new avenues for promoting long-term health starting from early life.
Collapse
Affiliation(s)
- Giulia Catassi
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168, Rome, Italy
- Pediatric Gastroenterology and Liver Unit, Umberto I Hospital, Sapienza University of Rome, Rome, Italy
| | - Sandra Garcia Mateo
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168, Rome, Italy
- Department of Gastroenterology, Lozano Blesa University Hospital, 50009, Zaragossa, Spain
| | - Annamaria Sara Occhionero
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168, Rome, Italy
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie Dell'Apparato DigerenteMedicina Interna E Gastroenterologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Chiara Esposito
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168, Rome, Italy
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie Dell'Apparato DigerenteMedicina Interna E Gastroenterologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Valentina Giorgio
- Department of Woman and Child Health and Public Health, UOC Pediatria, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Marina Aloi
- Pediatric Gastroenterology and Liver Unit, Umberto I Hospital, Sapienza University of Rome, Rome, Italy
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168, Rome, Italy
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie Dell'Apparato DigerenteMedicina Interna E Gastroenterologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giovanni Cammarota
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168, Rome, Italy
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie Dell'Apparato DigerenteMedicina Interna E Gastroenterologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Gianluca Ianiro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168, Rome, Italy.
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie Dell'Apparato DigerenteMedicina Interna E Gastroenterologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| |
Collapse
|
15
|
Frutos-Grilo E, Ana Y, Gonzalez-de Miguel J, Cardona-I-Collado M, Rodriguez-Arce I, Serrano L. Bacterial live therapeutics for human diseases. Mol Syst Biol 2024; 20:1261-1281. [PMID: 39443745 PMCID: PMC11612307 DOI: 10.1038/s44320-024-00067-0] [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: 06/13/2024] [Revised: 08/19/2024] [Accepted: 09/12/2024] [Indexed: 10/25/2024] Open
Abstract
The genomic revolution has fueled rapid progress in synthetic and systems biology, opening up new possibilities for using live biotherapeutic products (LBP) to treat, attenuate or prevent human diseases. Among LBP, bacteria-based therapies are particularly promising due to their ability to colonize diverse human tissues, modulate the immune system and secrete or deliver complex biological products. These bacterial LBP include engineered pathogenic species designed to target specific diseases, and microbiota species that promote microbial balance and immune system homeostasis, either through local administration or the gut-body axes. This review focuses on recent advancements in preclinical and clinical trials of bacteria-based LBP, highlighting both on-site and long-reaching strategies.
Collapse
Affiliation(s)
- Elisabet Frutos-Grilo
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Yamile Ana
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Javier Gonzalez-de Miguel
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Marcel Cardona-I-Collado
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Irene Rodriguez-Arce
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain.
| | - Luis Serrano
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain.
- ICREA, Pg. Lluis Companys 23, Barcelona, Spain.
| |
Collapse
|
16
|
Amar Y, Grube J, Köberle M, Schaubeck M, Biedermann T, Volz T. Bifidobacterium breve DSM 32583 and Limosilactobacillus fermentum CECT5716 postbiotics attenuate S. aureus and IL-33-induced Th2 responses. Microbiol Res 2024; 289:127913. [PMID: 39316930 DOI: 10.1016/j.micres.2024.127913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/04/2024] [Accepted: 09/15/2024] [Indexed: 09/26/2024]
Abstract
Over the past decades, the prevalence of allergic diseases noticeably increased in industrialized countries. The Th2 immune response plays a central role in these pathologies and its modulation using pro-/postbiotics constitutes a promising approach to prevent or alleviate disease symptoms. The aim of this in vitro study, was to investigate the ability of human milk-derived Bifidobacterium breve DSM 32583 (Bb) and Limosilactobacillus fermentum CECT5716 (Lf), to modulate the Th2 induced responses. To this end, Th2 cells were generated by co-culturing of human naïve Th cells with monocyte-derived dendritic cells (moDCs) either stimulated with Staphylococcus aureus or IL-33. The immunomodulatory effects of pro-/postbiotic preparations of Bb and Lf on moDCs and Th2 cells were evaluated in terms of maturation markers expression and cytokines production. Remarkably, the tested strains induced the anti-inflammatory cytokine IL-10 in moDCs, in a strain-, dose- and viability-dependent manner with no significant upregulation of IL-12p70 nor CD83, CD86 or HLA-DR. Interestingly, Bb and Lf postbiotics were able to dampen the Th2/Th1 response induced upon S. aureus- or IL-33 stimulation. They were also able to synergistically induce IL-10 in moDCs and T cells, upon co-stimulation with LPS. Finally, we observed that live probiotics triggered a mild Th1 response that was attenuated in the presence of galacto-oligosaccharides. Altogether, Bb and Lf pro-/postbiotics exhibited remarkable immune regulatory effects on both moDCs and Th2 cells. Therefore, further in vivo studies should be considered to validate these findings and assess their ability to prevent allergy or alleviate its symptoms in affected patients.
Collapse
Affiliation(s)
- Yacine Amar
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich 80802, Germany.
| | - Jana Grube
- HiPP GmbH & Co. Vertrieb KG, Pfaffenhofen (Ilm) 85276, Germany
| | - Martin Köberle
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich 80802, Germany
| | | | - Tilo Biedermann
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich 80802, Germany
| | - Thomas Volz
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich 80802, Germany
| |
Collapse
|
17
|
Bouillon GA, Xie Z, Nielsen DS, Wiese M, Nauta A. Propionate Production by Infant Fecal Microbiota Is Inversely Correlated with the Protein Glycation Level of Supplemented Infant Formula Ex Vivo. Nutrients 2024; 16:4047. [PMID: 39683441 DOI: 10.3390/nu16234047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES After birth, mothers provide the best nutrition for the healthy growth and development of their infants and the developing gut microbiota through breastfeeding. When breastfeeding is not or insufficiently available, infant formula is the only safe alternative. The production of infant formula includes heat-processing, which may induce protein glycation. Protein glycation has been shown to reduce protein digestion and absorption. The reduction in protein digestion and absorption because of protein glycation has been speculated to also impact gut comfort parameters as well as overnight sleep. METHODS As this could be partially due to the effect on the bacteria that reside in the infant's gastrointestinal tract, we investigated whether protein glycation in infant formula impacts the composition and activity of infant gut microbiota by performing an in vitro study using the CoMiniGut colon model and fecal inocula obtained from a healthy six-month-old term infant. Incubations were performed for 24 h using a predigested infant formula-supplemented medium with varying levels of glycation (6.5-44.5%). RESULTS Our data indicate that high protein glycation increases microbial diversity and the relative abundance of Clostridium neonatale from 6.4% of the inoculum to around 25.5% of 20.8% glycation. Interestingly, propionate levels were inversely correlated with protein glycation levels after 24 h of incubation, with the 44.5% blocked lysine sample giving rise to 60% lower propionate levels as compared to the 6.4% sample. Higher propionate levels have been linked with longer uninterrupted sleep overnight, which could be indicative of the underlying mechanism of reduced crying/fussy time during nights for infants fed with a formula containing lower amounts of glycated protein.
Collapse
Affiliation(s)
- Grégoire A Bouillon
- Section for Food Microbiology, Gut Health and Fermentation, Department of Food Science, University of Copenhagen, Rolighedsvej 26, DK-1958 Frederiksberg, Denmark
| | - Zhuqing Xie
- Section for Food Microbiology, Gut Health and Fermentation, Department of Food Science, University of Copenhagen, Rolighedsvej 26, DK-1958 Frederiksberg, Denmark
| | - Dennis S Nielsen
- Section for Food Microbiology, Gut Health and Fermentation, Department of Food Science, University of Copenhagen, Rolighedsvej 26, DK-1958 Frederiksberg, Denmark
| | - Maria Wiese
- Section for Food Microbiology, Gut Health and Fermentation, Department of Food Science, University of Copenhagen, Rolighedsvej 26, DK-1958 Frederiksberg, Denmark
| | - Arjen Nauta
- FrieslandCampina, Stationsplein 4, 3818 LE Amersfoort, The Netherlands
| |
Collapse
|
18
|
Yang R, Shi Z, Li Y, Huang X, Li Y, Li X, Chen Q, Hu Y, Li X. Research focus and emerging trends of the gut microbiome and infant: a bibliometric analysis from 2004 to 2024. Front Microbiol 2024; 15:1459867. [PMID: 39633813 PMCID: PMC11615055 DOI: 10.3389/fmicb.2024.1459867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024] Open
Abstract
Background Over the past two decades, gut microbiota has demonstrated unprecedented potential in human diseases and health. The gut microbiota in early life is crucial for later health outcomes. This study aims to reveal the knowledge collaboration network, research hotspots, and explore the emerging trends in the fields of infant and gut microbiome using bibliometric analysis. Method We searched the literature on infant and gut microbiome in the Web of Science Core Collection (WOSCC) database from 2004 to 2024. CiteSpace V (version: 6.3.R1) and VOSview (version: 1.6.20) were used to display the top authors, journals, institutions, countries, authors, keywords, co-cited articles, and potential trends. Results A total of 9,899 documents were retrieved from the Web of Science Core Collection. The United States, China, and Italy were the three most productive countries with 3,163, 1,510, and 660 publications. The University of California System was the most prolific institution (524 publications). Van Sinderen, Douwe from University College Cork of Ireland was the most impactful author. Many studies have focused on atopic dermatitis (AD), necrotizing enterocolitis (NEC), as well as the immune mechanisms and microbial treatments for these diseases, such as probiotic strains mixtures and human milk oligosaccharides (HMOs). The mother-to-infant microbiome transmission, chain fatty acids, and butyrate maybe the emerging trends. Conclusion This study provided an overview of the knowledge structure of infant and gut microbiome, as well as a reference for future research.
Collapse
Affiliation(s)
- Ru Yang
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Zeyao Shi
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Yuan Li
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Xi Huang
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Yingxin Li
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Xia Li
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Qiong Chen
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Yanling Hu
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Xiaowen Li
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| |
Collapse
|
19
|
Froń A, Orczyk-Pawiłowicz M. Breastfeeding Beyond Six Months: Evidence of Child Health Benefits. Nutrients 2024; 16:3891. [PMID: 39599677 PMCID: PMC11597163 DOI: 10.3390/nu16223891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/05/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
Breastfeeding is globally recognized as the optimal method of infant nutrition, offering health benefits for both the child and the mother, making it a public health priority. However, the potential advantages of breastfeeding extend well beyond initial months. Breast milk adapts to the evolving needs of the growing infant, and its immunological, microbiological, and biochemical properties have been associated with enhanced protection against infections and chronic diseases, improved growth and development, and lower rates of hospitalization and mortality. This review explores the evidence supporting the continuation of breastfeeding beyond six months. More meticulous studies employing consistent methodologies and addressing confounders are essential. This will enable a more accurate determination of the extent and mechanisms of the positive impact of prolonged breastfeeding and allow for the implementation of effective public health strategies.
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;
| | | |
Collapse
|
20
|
Ma X, Mo J, Shi L, Cheng Y, Feng J, Qin J, Su W, Lv J, Li S, Li Q, Tan H, Han B. Isolation and characterization of Bifidobacterium spp. from breast milk with different human milk oligosaccharides utilization and anti-inflammatory capacity. Food Res Int 2024; 196:115092. [PMID: 39614508 DOI: 10.1016/j.foodres.2024.115092] [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: 06/24/2024] [Revised: 09/10/2024] [Accepted: 09/12/2024] [Indexed: 12/01/2024]
Abstract
Breast milk is the best source of nutrition for infants. Human milk oligosaccharides (HMOs) and the corresponding HMOs-consuming Bifidobacterium positively influence infant health. This study aims to isolate and characterize Bifidobacterium from breast milk of healthy Chinese mothers, identifying the most efficacious strains for inclusion in simulated maternal milk formulas. Nine Bifidobacterium strains (two of B. breve and seven of B. infantis) were isolated, exhibiting a broad spectrum of probiotic potential. This included tolerance to simulated infant gastrointestinal conditions, notable adhesion, antibacterial, antioxidant activities, and HMOs utilization ability. Lacto-N-Tetraose (LNT) is preferred in early growth among Bifidobacterium isolates. B. breve showed a preference for LNT, whereas B. infantis showed a preference for fucosylated HMOs, and displayed reduced utilization of sialylated HMOs. They also exhibited robust safety profiles, including no hemolytic activity, an appropriate D/L lactate-producing ratio, and non-toxicity in an acute oral toxicity assay on mice. It is noteworthy that B. breve N-90, O-147, B. infantis O-161 and R-1 exhibited anti-inflammatory effects in LPS-induced RAW 264.7 cells. Specifically, a notable reduction in TNF-α levels was observed in pre-treatment, while a decrease in IL-1β and IL-6 levels in co-treatment. B. breve N-90 and B. infantis R-1 were identified finally as promising probiotic candidates. Their whole-genome sequencing analysis confirmed presence of functional genes associated with gastrointestinal colonization, antioxidation, and glycoside hydrolase activity on HMOs. The annotation for antibiotic resistance and virulence genes concurred with phenotypes, further validating the safety. Breast milk is a good source for Bifidobacteria isolation, while Bifidobacteria utilize HMOs in a strain-dependent manner. The two selected strains, B. breve N-90 and B. infantis R-1, are potential candidates for inclusion in simulated maternal milk formulas and deserved further in vivo investigation for their health-promoting effects.
Collapse
Affiliation(s)
- Xinxin Ma
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jianhui Mo
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Lu Shi
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yue Cheng
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jiayu Feng
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jiale Qin
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Wanghong Su
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jia Lv
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, Shaanxi 710061, China
| | - Shaoru Li
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, Shaanxi 710061, China
| | - Qiang Li
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, Shaanxi 710061, China
| | - Hui Tan
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| | - Bei Han
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
21
|
van Duuren IC, van Hengel ORJ, Penders J, Duijts L, Smits HH, Tramper-Stranders GA. The developing immune system in preterm born infants: From contributor to potential solution for respiratory tract infections and wheezing. Allergy 2024; 79:2924-2942. [PMID: 39382056 DOI: 10.1111/all.16342] [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: 06/07/2024] [Revised: 09/05/2024] [Accepted: 09/17/2024] [Indexed: 10/10/2024]
Abstract
Moderate-late preterm-born infants experience more frequent and severe respiratory tract infections and wheezing compared to term-born infants. Decreasing the risk on respiratory tract infections and wheezing in this group is vital to improve quality of life and reduce medical consumption during infancy, but also to reduce the risk on asthma and COPD later in life. Until now, moderate-late preterm infants are underrepresented in research and mechanisms underlying their morbidity are largely unknown, although they represent 80% of all preterm-born infants. In order to protect these infants effectively, it is essential to understand the role of the immune system in early life respiratory health and to identify strategies to optimize immune development and respiratory health. This review elaborates on risk factors and preventative measures concerning respiratory tract infections and wheezing in preterm-born infants, exploring their impact on the immune system and microbiome. Factors discussed are early life antibiotic use, birth mode, feeding type and living environment. Further, differences in adaptive and innate immune maturation between term and preterm infants are discussed, as well as differences in local immune reactions in the lungs. Finally, preventative strategies are being explored, including microbiota transplantation, immune modulation (through pre-, pro-, syn- and postbiotics, bacterial lysates, vaccinations, and monoclonal antibodies) and antibiotic prophylaxis.
Collapse
Affiliation(s)
- Inger C van Duuren
- Department of Paediatrics, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Sophia Children's Hospital - Erasmus MC, Rotterdam, The Netherlands
| | - Oscar R J van Hengel
- Leiden University Center of Infectious Disease (LU-CID), Leiden, The Netherlands
| | - John Penders
- Department of Medical Microbiology, Infectious Diseases and Infection Prevention, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Liesbeth Duijts
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Sophia Children's Hospital - Erasmus MC, Rotterdam, The Netherlands
| | - Hermelijn H Smits
- Leiden University Center of Infectious Disease (LU-CID), Leiden, The Netherlands
| | - Gerdien A Tramper-Stranders
- Department of Paediatrics, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
- Department of Neonatal and Pediatric Intensive Care, Division of Neonatology, Sophia Children's Hospital, ErasmusMC, Rotterdam, The Netherlands
| |
Collapse
|
22
|
Espín JC, Jarrín‐Orozco MP, Osuna‐Galisteo L, Ávila‐Gálvez MÁ, Romo‐Vaquero M, Selma MV. Perspective on the Coevolutionary Role of Host and Gut Microbiota in Polyphenol Health Effects: Metabotypes and Precision Health. Mol Nutr Food Res 2024; 68:e2400526. [PMID: 39538982 PMCID: PMC11605795 DOI: 10.1002/mnfr.202400526] [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: 07/08/2024] [Revised: 09/25/2024] [Indexed: 11/16/2024]
Abstract
"Personalized nutrition" aims to establish nutritional strategies to improve health outcomes for non-responders. However, it is utopian since most people share similar nutritional requirements. "Precision health," encompassing lifestyles, may be more fitting. Dietary (poly)phenols are "healthy" but non-nutritional molecules (thus, we can live without them). The gut microbiota influences (poly)phenol effects, producing metabolites with different activity than their precursors. Furthermore, producing distinctive metabolites, like urolithins, lunularin, and equol, leads to the term "polyphenol-related gut microbiota metabotypes," grouping individuals based on a genuine microbial metabolism of ellagic acid, resveratrol, and isoflavones, respectively. Additionally, (poly)phenols exert prebiotic-like effects through their antimicrobial activities, typically reducing microbial diversity and modulating microbiota functionality by impacting its composition and transcriptomics. Since the gut microbiota perceives (poly)phenols as a threat, (poly)phenol effects are mostly a consequence of microbiota adaptation through differential (poly)phenol metabolism (e.g., distinctive reductions, dehydroxylations, etc.). This viewpoint is less prosaic than considering (poly)phenols as essential nutritional players in human health, yet underscores their health significance in a coevolutionary partnership with the gut microbiota. In the perspective on the gut microbiota and (poly)phenols interplay, microbiota metabotypes could arbiter health effects. An innovative aspect is also emphasized: modulating the interacting microbial networks without altering the composition.
Collapse
Affiliation(s)
- Juan Carlos Espín
- Laboratory of Food & Health; Research Group on Quality, Safety and Bioactivity of Plant FoodsCEBAS‐CSIC30100 Campus de EspinardoMurciaSpain
| | - María Paula Jarrín‐Orozco
- Laboratory of Food & Health; Research Group on Quality, Safety and Bioactivity of Plant FoodsCEBAS‐CSIC30100 Campus de EspinardoMurciaSpain
| | - Leire Osuna‐Galisteo
- Laboratory of Food & Health; Research Group on Quality, Safety and Bioactivity of Plant FoodsCEBAS‐CSIC30100 Campus de EspinardoMurciaSpain
| | - María Ángeles Ávila‐Gálvez
- Laboratory of Food & Health; Research Group on Quality, Safety and Bioactivity of Plant FoodsCEBAS‐CSIC30100 Campus de EspinardoMurciaSpain
| | - María Romo‐Vaquero
- Laboratory of Food & Health; Research Group on Quality, Safety and Bioactivity of Plant FoodsCEBAS‐CSIC30100 Campus de EspinardoMurciaSpain
| | - María Victoria Selma
- Laboratory of Food & Health; Research Group on Quality, Safety and Bioactivity of Plant FoodsCEBAS‐CSIC30100 Campus de EspinardoMurciaSpain
| |
Collapse
|
23
|
Yin X, Shi Y, Sheng T, Ji C. Early-Life Gut Microbiota: A Possible Link Between Maternal Exposure to Non-Nutritive Sweeteners and Metabolic Syndrome in Offspring. Nutr Rev 2024:nuae140. [PMID: 39348276 DOI: 10.1093/nutrit/nuae140] [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] [Indexed: 10/02/2024] Open
Abstract
Metabolic syndrome (MetS) is recognized as a group of metabolic abnormalities, characterized by clustered interconnected traits that elevate the risks of obesity, cardiovascular and atherosclerotic diseases, hyperlipidemia, and type 2 diabetes mellitus. Non-nutritive sweeteners (NNS) are commonly consumed by those with imbalanced calorie intake, especially in the perinatal period. In the past, accumulating evidence showed the transgenerational and mediated roles of human microbiota in the development of early-life MetS. Maternal exposure to NNS has been recognized as a risk factor for filial metabolic disturbance through various mechanisms, among which gut microbiota and derived metabolites function as nodes linking NNS and MetS in early life. Despite the widespread consumption of NNS, there remain growing concerns about their transgenerational impact on metabolic health. There is growing evidence of NNS being implicated in the development of metabolic abnormalities. Intricate complexities exist and a comprehensive understanding of how the gut microbiota interacts with mechanisms related to maternal NNS intake and disrupts metabolic homeostasis of offspring is critical to realize its full potential in preventing early-life MetS. This review aims to elucidate the effects of early-life gut microbiota and links to maternal NNS exposure and imbalanced offspring metabolic homeostasis and discusses potential perspectives and challenges, which may provide enlightenment and understanding into optimal perinatal nutritional management.
Collapse
Affiliation(s)
- Xiaoxiao Yin
- Nanjing Medical Research Center for Women and Children, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu 210004, China
- School of Nursing, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yujie Shi
- Nanjing Medical Research Center for Women and Children, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu 210004, China
| | - Tongtong Sheng
- School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Chenbo Ji
- Nanjing Medical Research Center for Women and Children, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu 210004, China
- School of Nursing, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
24
|
Azanaw J, Malede A, Yalew HF, Worede EA. Determinants of diarrhoeal diseases among under-five children in Africa (2013-2023): a comprehensive systematic review highlighting geographic variances, socioeconomic influences, and environmental factors. BMC Public Health 2024; 24:2399. [PMID: 39232730 PMCID: PMC11373296 DOI: 10.1186/s12889-024-19962-0] [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: 02/22/2024] [Accepted: 09/02/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND Diarrhea diseases continue to present a significant threat to the well-being of children under the age of five in Africa, thereby contributing substantially to both morbidity and mortality rates. The period spanning between January 2013 and December 2023 has witnessed persistent challenges in the fight against these diseases, thereby necessitating a thorough investigation into the factors that determine their occurrence. It is important to note that the burden of diarrhea diseases is not evenly distributed across the continent, with residence, socioeconomic, and environmental factors playing pivotal roles in shaping the prevalence and incidence rates. Consequently, this systematic review aimed to consolidate and analyze the existing body of literature on the determinants of diarrhea diseases among children under the age of five in Africa between January 2013 and December 2023. METHOD The systematic review employed a rigorous methodological approach to examine the determinants of diarrhea diseases among children under the age of five in Africa between January 2013 and December 2023. A comprehensive search strategy was implemented, utilizing databases such as PubMed, Scopus, and Web of Science, and incorporating relevant keywords. The inclusion criteria focused on studies published within the specified timeframe, with a specific focus on the determinants of diarrhea disease among children under the age of five in Africa. The study selection process involved a two-stage screening, with independent reviewers evaluating titles, abstracts, and full texts to determine eligibility. The quality assessment, employing a standardized tool, ensured the inclusion of studies with robust methodologies. Data extraction encompassed key study details, including demographics, residence factors, socioeconomic influences, environmental variables, and intervention outcomes. RESULTS The search yielded a total of 12,580 articles across 25 African countries; however, only 97 of these articles met the inclusion criteria and were ultimately included in the systematic review. The systematic review revealed geographic and seasonal disparities in the prevalence of diarrhoeal diseases across different countries in Africa. Factors such as age-related vulnerabilities, gender disparities, maternal occupation, disposal of young children's stools, and economic status were identified as significant determinants of the prevalence of diarrhea disease. CONCLUSION This systematic review provides a comprehensive understanding of the determinants of diarrhea diseases among children under the age of five in Africa between January 2013 and December 2023. The nuanced analysis of residence variations, socioeconomic influences, environmental factors, and intervention outcomes underscores the complex nature of this issue. The findings highlight the necessity for region-specific and context-sensitive interventions to address the unique challenges faced by diverse communities. This review serves as a valuable resource for policymakers, healthcare professionals, and researchers, guiding the development of evidence-based strategies aimed at reducing the burden of diarrhea diseases and improving child health outcomes in Africa.
Collapse
Affiliation(s)
- Jember Azanaw
- Department of Environmental and Occupational Health and Safety, Institute of Public Health, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia.
| | - Asmamaw Malede
- Department of Environmental and Occupational Health and Safety, Institute of Public Health, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Hailemariam Feleke Yalew
- Department of Environmental and Occupational Health and Safety, Institute of Public Health, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Eshetu Abera Worede
- Department of Environmental and Occupational Health and Safety, Institute of Public Health, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
25
|
Tripp P, Davis EC, Gurung M, Rosa F, Bode L, Fox R, LeRoith T, Simecka C, Seppo AE, Järvinen KM, Yeruva L. Infant Microbiota Communities and Human Milk Oligosaccharide Supplementation Independently and Synergistically Shape Metabolite Production and Immune Responses in Healthy Mice. J Nutr 2024; 154:2871-2886. [PMID: 39069270 PMCID: PMC11393170 DOI: 10.1016/j.tjnut.2024.07.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/03/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND Multiple studies have demonstrated associations between the early-life gut microbiome and incidence of inflammatory and autoimmune disease in childhood. Although microbial colonization is necessary for proper immune education, it is not well understood at a mechanistic level how specific communities of bacteria promote immune maturation or drive immune dysfunction in infancy. OBJECTIVES In this study, we aimed to assess whether infant microbial communities with different overall structures differentially influence immune and gastrointestinal development in healthy mice. METHODS Germ-free mice were inoculated with fecal slurries from Bifidobacterium longum subspecies infantis positive (BIP) or B. longum subspecies infantis negative (BIN) breastfed infants; half of the mice in each group were also supplemented with a pool of human milk oligosaccharides (HMOs) for 14 d. Cecal microbiome composition and metabolite production, systemic and mucosal immune outcomes, and intestinal morphology were assessed at the end of the study. RESULTS The results showed that inoculation with a BIP microbiome results in a remarkably distinct microbial community characterized by higher relative abundances of cecal Clostridium senu stricto, Ruminococcus gnavus, Cellulosilyticum sp., and Erysipelatoclostridium sp. The BIP microbiome produced 2-fold higher concentrations of cecal butyrate, promoted branched short-chain fatty acid (SCFA) production, and further modulated serotonin, kynurenine, and indole metabolism relative to BIN mice. Further, the BIP microbiome increased the proportions of innate and adaptive immune cells in spleen, while HMO supplementation increased proliferation of mesenteric lymph node cells to phorbol myristate acetate and lipopolysaccharide and increased serum IgA and IgG concentrations. CONCLUSIONS Different microbiome compositions and HMO supplementation can modulate SCFA and tryptophan metabolism and innate and adaptive immunity in young, healthy mice, with potentially important implications for early childhood health.
Collapse
Affiliation(s)
- Patricia Tripp
- USDA-ARS, SEA, Microbiome and Metabolism Research Unit, Arkansas Children's Nutrition Center, Little Rock, AR, United States
| | - Erin C Davis
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, NY, United States
| | - Manoj Gurung
- USDA-ARS, SEA, Microbiome and Metabolism Research Unit, Arkansas Children's Nutrition Center, Little Rock, AR, United States
| | - Fernanda Rosa
- School of Veterinary Medicine, Texas Tech University, Amarillo, TX, United States
| | - Lars Bode
- Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence, University of California San Diego, La Jolla, CA, United States; Department of Pediatrics, University of California San Diego, La Jolla, CA, United States
| | - Renee Fox
- USDA-ARS, SEA, Microbiome and Metabolism Research Unit, Arkansas Children's Nutrition Center, Little Rock, AR, United States
| | - Tanya LeRoith
- Department of Biomedical Sciences & Pathobiology, Virginia Tech, Blacksburg, VA, United States
| | - Christy Simecka
- Division of Laboratory Animal Medicine University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Antti E Seppo
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, NY, United States
| | - Kirsi M Järvinen
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, NY, United States; Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States.
| | - Laxmi Yeruva
- USDA-ARS, SEA, Microbiome and Metabolism Research Unit, Arkansas Children's Nutrition Center, Little Rock, AR, United States.
| |
Collapse
|
26
|
Davis EC, Monaco CL, Insel R, Järvinen KM. Gut microbiome in the first 1000 days and risk for childhood food allergy. Ann Allergy Asthma Immunol 2024; 133:252-261. [PMID: 38494114 PMCID: PMC11344696 DOI: 10.1016/j.anai.2024.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/11/2024] [Accepted: 03/11/2024] [Indexed: 03/19/2024]
Abstract
OBJECTIVE To summarize recent data on the association between gut microbiome composition and food allergy (FA) in early childhood and highlight potential host-microbiome interactions that reinforce or abrogate oral tolerance. DATA SOURCES PubMed search of English-language articles related to FA, other atopic disease, and the gut microbiome in pregnancy and early childhood. STUDY SELECTIONS Human studies published after 2015 assessing the relationship between the gut bacteriome and virome in the first 2 years of life and FA or food sensitization development in early childhood were prioritized. Additional human studies conducted on the prenatal gut microbiome or other atopic diseases and preclinical studies are also discussed. RESULTS Children who developed FA harbored lower abundances of Bifidobacterium and Clostridia species and had a less mature microbiome during infancy. The early bacterial microbiome protects against FA through production of anti-inflammatory metabolites and induction of T regulatory cells and may also affect FA risk through a role in trained immunity. Infant enteric phage communities are related to childhood asthma development, though no data are available for FA. Maternal gut microbiome during pregnancy is associated with childhood FA risk, potentially through transplacental delivery of maternal bacterial metabolites, though human studies are lacking. CONCLUSION The maternal and infant microbiomes throughout the first 1000 days of life influence FA risk through a number of proposed mechanisms. Further large, longitudinal cohort studies using taxonomic, functional, and metabolomic analysis of the bacterial and viral microbiomes are needed to provide further insight on the host-microbe interactions underlying FA pathogenesis in childhood.
Collapse
Affiliation(s)
- Erin C Davis
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, New York
| | - Cynthia L Monaco
- Division of Infectious Disease, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York; Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Richard Insel
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, New York
| | - Kirsi M Järvinen
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, New York; Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York; Division of Allergy, Immunology, and Rheumatology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York.
| |
Collapse
|
27
|
Herrera-Quintana L, Vázquez-Lorente H, Hinojosa-Nogueira D, Plaza-Diaz J. Relationship between Infant Feeding and the Microbiome: Implications for Allergies and Food Intolerances. CHILDREN (BASEL, SWITZERLAND) 2024; 11:1030. [PMID: 39201963 PMCID: PMC11353207 DOI: 10.3390/children11081030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/16/2024] [Accepted: 08/20/2024] [Indexed: 09/03/2024]
Abstract
Childhood is a critical period for immune system development, which is greatly influenced by the gut microbiome. Likewise, a number of factors affect the gut microbiome composition and diversity, including breastfeeding, formula feeding, and solid foods introduction. In this regard, several studies have previously demonstrated that breastfeeding promotes a favorable microbiome. In contrast, formula feeding and the early incorporation of certain solid foods may adversely affect microbiome development. Additionally, there is increasing evidence that disruptions in the early microbiome can lead to allergic conditions and food intolerances. Thus, developing strategies to promote optimal infant nutrition requires an understanding of the relationship between infant nutrition and long-term health. The present review aims to examine the relationship between infant feeding practices and the microbiome, as well as its implications on allergies and food intolerances in infants. Moreover, this study synthesizes existing evidence on how different eating habits influence the microbiome. It highlights their implications for the prevention of allergies and food intolerances. In conclusion, introducing allergenic solid foods before six months, alongside breastfeeding, may significantly reduce allergies and food intolerances risks, being also associated with variations in gut microbiome and related complications.
Collapse
Affiliation(s)
- Lourdes Herrera-Quintana
- Department of Physiology, Schools of Pharmacy and Medicine, University of Granada, 18071 Granada, Spain; (L.H.-Q.); (H.V.-L.)
- Biomedical Research Center, Health Sciences Technology Park, University of Granada, 18016 Granada, Spain
| | - Héctor Vázquez-Lorente
- Department of Physiology, Schools of Pharmacy and Medicine, University of Granada, 18071 Granada, Spain; (L.H.-Q.); (H.V.-L.)
- Biomedical Research Center, Health Sciences Technology Park, University of Granada, 18016 Granada, Spain
| | - Daniel Hinojosa-Nogueira
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Laboratorio del Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario de Málaga (Virgen de la Victoria), 29590 Málaga, Spain;
| | - Julio Plaza-Diaz
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, Campus de Cartuja s/n, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria IBS, GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
| |
Collapse
|
28
|
Ottria R, Xynomilakis O, Casati S, Ciuffreda P. Pre- to Postbiotics: The Beneficial Roles of Pediatric Dysbiosis Associated with Inflammatory Bowel Diseases. Microorganisms 2024; 12:1582. [PMID: 39203424 PMCID: PMC11356122 DOI: 10.3390/microorganisms12081582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 09/03/2024] Open
Abstract
Probiotics are "live microorganisms which, when administered in adequate amount, confer health benefits on the host". They can be found in certain foods like yogurt and kefir and in dietary supplements. The introduction of bacterial derivatives has not only contributed to disease control but has also exhibited promising outcomes, such as improved survival rates, immune enhancement, and growth promotion effects. It is interesting to note that the efficacy of probiotics goes beyond the viability of the bacteria, giving rise to concepts like paraprobiotics, non-viable forms of probiotics, and postbiotics. Paraprobiotics offer various health benefits in children with intestinal dysbiosis, contributing to improved digestive health, immune function, and overall well-being. In this review, the potential of these therapeutic applications as alternatives to pharmacological agents for treating pediatric intestinal dysbiosis will be thoroughly evaluated. This includes an analysis of their efficacy, safety, long-term benefits, and their ability to restore gut microbiota balance, improve digestive health, enhance immune function, and reduce inflammation. The aim is to determine if these non-pharmacological interventions can effectively and safely manage intestinal dysbiosis in children, reducing the need for conventional medications and their side effects.
Collapse
Affiliation(s)
- Roberta Ottria
- Dipartimento di Scienze Biomediche e Cliniche, Università degli Studi di Milano, 20157 Milan, Italy; (O.X.); (S.C.); (P.C.)
| | | | | | | |
Collapse
|
29
|
Houngbédji M, Jespersen JS, Wilfrid Padonou S, Jespersen L. Cereal-based fermented foods as microbiota-directed products for improved child nutrition and health in sub-Saharan Africa. Crit Rev Food Sci Nutr 2024:1-22. [PMID: 38973125 DOI: 10.1080/10408398.2024.2365342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2024]
Abstract
Several strategies, programs and policies have long been developed and implemented to alleviate child malnutrition in sub-Saharan African countries. However, stunting and wasting still persist at an alarming rate, suggesting that alternative strategies are needed to induce faster progress toward the 2030 SDGs targets of reducing malnutrition. Gut microbiota-directed intervention is now being recognized as an unconventional powerful approach to mitigate malnutrition and improve overall child health. In an African setting, manufactured probiotic and synbiotic foods or supplements may not be successful owing to the non-affordability and high attachment of African populations to their food tradition. This review analyses the potential of indigenous fermented cereal-based products including porridges, doughs, beverages, bread- and yoghurt-like products, to be used as microbiota-directed foods for over 6 months children. The discussion includes relevant strategies to effectively enhance the beneficial effects of these products on gut microbiota composition for improved child health and nutrition in sub-Saharan Africa. Characterization of probiotic features and general safety of food processing in sub-Saharan Africa as well as randomized clinical studies are still lacking to fully ascertain the health effects and suitability of these fermented foods in preventing and treating child malnutrition and diarrhea.
Collapse
Affiliation(s)
- Marcel Houngbédji
- Laboratoire de Sciences et Technologies des Aliments, Faculté des Sciences Agronomiques, Université d'Abomey-Calavi, Jéricho, Cotonou, Benin
- Laboratoire de Sciences et Technologie des Aliments, des Bioressources et de Nutrition Humaine, Université Nationale d'Agriculture, Sakété, Bénin
| | | | - Sègla Wilfrid Padonou
- Laboratoire de Sciences et Technologies des Aliments, Faculté des Sciences Agronomiques, Université d'Abomey-Calavi, Jéricho, Cotonou, Benin
- Laboratoire de Sciences et Technologie des Aliments, des Bioressources et de Nutrition Humaine, Université Nationale d'Agriculture, Sakété, Bénin
| | - Lene Jespersen
- Department of Food Science, University of Copenhagen, Copenhagen, Frederiksberg C, Denmark
| |
Collapse
|
30
|
Nobs SP, Elinav E. Complement(ing) the microbiome in infants through breastmilk. Cell Res 2024; 34:469-470. [PMID: 38443564 PMCID: PMC11217263 DOI: 10.1038/s41422-024-00944-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024] Open
Affiliation(s)
- Samuel P Nobs
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Elinav
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel.
- Microbiome & Cancer Division, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
31
|
Selma-Royo M, Dubois L, Manara S, Armanini F, Cabrera-Rubio R, Valles-Colomer M, González S, Parra-Llorca A, Escuriet R, Bode L, Martínez-Costa C, Segata N, Collado MC. Birthmode and environment-dependent microbiota transmission dynamics are complemented by breastfeeding during the first year. Cell Host Microbe 2024; 32:996-1010.e4. [PMID: 38870906 PMCID: PMC11183301 DOI: 10.1016/j.chom.2024.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/24/2024] [Accepted: 05/10/2024] [Indexed: 06/15/2024]
Abstract
The composition and maturation of the early-life microbiota are modulated by a number of perinatal factors, whose interplay in relation to microbial vertical transmission remains inadequately elucidated. Using recent strain-tracking methodologies, we analyzed mother-to-infant microbiota transmission in two different birth environments: hospital-born (vaginal/cesarean) and home-born (vaginal) infants and their mothers. While delivery mode primarily explains initial compositional differences, place of birth impacts transmission timing-being early in homebirths and delayed in cesarean deliveries. Transmission patterns vary greatly across species and birth groups, yet certain species, like Bifidobacterium longum, are consistently vertically transmitted regardless of delivery setting. Strain-level analysis of B. longum highlights relevant and consistent subspecies replacement patterns mainly explained by breastfeeding practices, which drive changes in human milk oligosaccharide (HMO) degrading capabilities. Our findings highlight how delivery setting, breastfeeding duration, and other lifestyle preferences collectively shape vertical transmission, impacting infant gut colonization during early life.
Collapse
Affiliation(s)
- Marta Selma-Royo
- Institute of Agrochemistry and Food Technology-Spanish National Research Council (IATA-CSIC), Paterna, Valencia, Spain; Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Léonard Dubois
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Serena Manara
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Federica Armanini
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Raúl Cabrera-Rubio
- Institute of Agrochemistry and Food Technology-Spanish National Research Council (IATA-CSIC), Paterna, Valencia, Spain
| | - Mireia Valles-Colomer
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy; MELIS Department, Universitat Pompeu Fabra, Barcelona, Spain
| | - Sonia González
- Department of Functional Biology, University of Oviedo, Oviedo, Spain; Diet Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Anna Parra-Llorca
- Health Research Institute La Fe, Neonatal Research Group, Division of Neonatology, Valencia, Spain
| | - Ramon Escuriet
- Gerencia de Procesos Integrales de Salud. Area Asistencial, Servicio Catalan de la Salud, Generalitat de Catalunya, Centre for Research in Health and Economics, Universidad Pompeu Fabra, Barcelona, Spain
| | - Lars Bode
- Department of Pediatrics, Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (LRF MOMI CORE), Human Milk Institute (HMI), University of California, San Diego, La Jolla, CA, USA
| | - Cecilia Martínez-Costa
- Department of Pediatrics, Hospital Clínico Universitario, University of Valencia, Spain; Nutrition Research Group of INCLIVA, Valencia, Spain
| | - Nicola Segata
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy.
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology-Spanish National Research Council (IATA-CSIC), Paterna, Valencia, Spain.
| |
Collapse
|
32
|
Wong CB, Huang H, Ning Y, Xiao J. Probiotics in the New Era of Human Milk Oligosaccharides (HMOs): HMO Utilization and Beneficial Effects of Bifidobacterium longum subsp. infantis M-63 on Infant Health. Microorganisms 2024; 12:1014. [PMID: 38792843 PMCID: PMC11124435 DOI: 10.3390/microorganisms12051014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
A healthy gut microbiome is crucial for the immune system and overall development of infants. Bifidobacterium has been known to be a predominant species in the infant gut; however, an emerging concern is the apparent loss of this genus, in particular, Bifidobacterium longum subsp. infantis (B. infantis) in the gut microbiome of infants in industrialized nations, underscoring the importance of restoring this beneficial bacterium. With the growing understanding of the gut microbiome, probiotics, especially infant-type human-residential bifidobacteria (HRB) strains like B. infantis, are gaining prominence for their unique ability to utilize HMOs and positively influence infant health. This article delves into the physiology of a probiotic strain, B. infantis M-63, its symbiotic relationship with HMOs, and its potential in improving gastrointestinal and allergic conditions in infants and children. Moreover, this article critically assesses the role of HMOs and the emerging trend of supplementing infant formulas with the prebiotic HMOs, which serve as fuel for beneficial gut bacteria, thereby emulating the protective effects of breastfeeding. The review highlights the potential of combining B. infantis M-63 with HMOs as a feasible strategy to improve health outcomes in infants and children, acknowledging the complexities and requirements for further research in this area.
Collapse
Affiliation(s)
- Chyn Boon Wong
- International Division, Morinaga Milk Industry Co., Ltd., 5-2, Higashi Shimbashi 1-Chome, Minato-ku, Tokyo 105-7122, Japan
| | - Huidong Huang
- Nutrition Research Institute, Junlebao Dairy Group Co., Ltd., 36 Shitong Road, Shijiazhuang 050221, China
| | - Yibing Ning
- Nutrition Research Institute, Junlebao Dairy Group Co., Ltd., 36 Shitong Road, Shijiazhuang 050221, China
| | - Jinzhong Xiao
- Morinaga Milk Industry (Shanghai) Co., Ltd., Room 509 Longemont Yes Tower, No. 369 Kaixuan Road, Changning District, Shanghai 200050, China
- Department of Microbiota Research, Graduate School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Research Center for Probiotics, Department of Nutrition and Health, China Agricultural University, Beijing 100093, China
| |
Collapse
|
33
|
Minami Y, Kanemura S, Kusaka J, Kinouchi M, Suzuki S, Iwasashi H, Nishino Y, Kakugawa Y, Miura K. Association between being breastfed in infancy and adult colorectal cancer risk among Japanese men and women. Sci Rep 2024; 14:9661. [PMID: 38671142 PMCID: PMC11053050 DOI: 10.1038/s41598-024-60448-1] [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: 12/06/2023] [Accepted: 04/23/2024] [Indexed: 04/28/2024] Open
Abstract
It has been postulated that being breastfed in infancy affects not only health status in childhood but also disease risk in adulthood. To investigate the association of being breastfed with the risks of adult colorectal cancer and benign tumor, we conducted a case-control study including 1190 colorectal cancer and 1585 benign tumor cases and 5301 controls, admitted to a single hospital in Miyagi Prefecture, Japan, between 1997 and 2013. History of having been breastfed was assessed using a self-administered questionnaire, and odds ratios (ORs) were estimated using unconditional logistic regression. There was no association between being breastfed and colorectal cancer risk (breastfed versus formula-only fed, OR = 1.21; 95% CI 0.87-1.67). There was also no association with the risk of benign tumor (OR = 1.04). On the other hand, analyses stratified by sex and birth year found heterogeneous associations. Women born after 1950 who had been breastfed tended to have increased risks of colorectal cancer (OR = 1.58) and benign tumor (OR = 1.51) relative to those who had been formula-only fed, although not statistically significant. In men born after 1950, being breastfed was associated with a significantly decreased risk of benign tumor (OR = 0.57; 95% CI 0.33-0.98).
Collapse
Affiliation(s)
- Yuko Minami
- Department of Health Sciences, Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan.
- Division of Cancer Epidemiology and Prevention, Miyagi Cancer Center Research Institute, 47-1 Nodayama, Medeshima-Shiode, Natori, Miyagi, 981-1293, Japan.
- Center for Preventive Medicine, Osaki Citizen Hospital, 2-3-15 Senjuji-Machi, Furukawa, Osaki, Miyagi, 989-6174, Japan.
| | - Seiki Kanemura
- Division of Cancer Epidemiology and Prevention, Miyagi Cancer Center Research Institute, 47-1 Nodayama, Medeshima-Shiode, Natori, Miyagi, 981-1293, Japan
| | - Jun Kusaka
- Department of Gastroenterology, Miyagi Cancer Center Hospital, 47-1 Nodayama, Medeshima-Shiode, Natori, Miyagi, 981-1293, Japan
| | - Makoto Kinouchi
- Department of Surgery, Miyagi Cancer Center Hospital, 47-1 Nodayama, Medeshima-Shiode, Natori, Miyagi, 981-1293, Japan
| | - Shinichi Suzuki
- Department of Gastroenterology, Miyagi Cancer Center Hospital, 47-1 Nodayama, Medeshima-Shiode, Natori, Miyagi, 981-1293, Japan
| | - Hajime Iwasashi
- Department of Surgery, Miyagi Cancer Center Hospital, 47-1 Nodayama, Medeshima-Shiode, Natori, Miyagi, 981-1293, Japan
| | - Yoshikazu Nishino
- Division of Cancer Epidemiology and Prevention, Miyagi Cancer Center Research Institute, 47-1 Nodayama, Medeshima-Shiode, Natori, Miyagi, 981-1293, Japan
- Deapartment of Epidemiology and Public Health, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
| | - Yoichiro Kakugawa
- Department of Surgery, Miyagi Cancer Center Hospital, 47-1 Nodayama, Medeshima-Shiode, Natori, Miyagi, 981-1293, Japan
- Department of Surgery, Sendai Red Cross Hospital, 2-43-3 Honcho, Yagiyama, Sendai, Miyagi, 982-8501, Japan
| | - Koh Miura
- Department of Surgery, Miyagi Cancer Center Hospital, 47-1 Nodayama, Medeshima-Shiode, Natori, Miyagi, 981-1293, Japan
- Department of Surgery, Omagari Kosei Medical Center, 8-65 Omagari-Toricho, Daisen, Akita, 014-0027, Japan
| |
Collapse
|
34
|
Zhang M, Zhou Y, Cui X, Zhu L. The Potential of Co-Evolution and Interactions of Gut Bacteria-Phages in Bamboo-Eating Pandas: Insights from Dietary Preference-Based Metagenomic Analysis. Microorganisms 2024; 12:713. [PMID: 38674657 PMCID: PMC11051890 DOI: 10.3390/microorganisms12040713] [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: 01/16/2024] [Revised: 03/26/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Bacteria and phages are two of the most abundant biological entities in the gut microbiome, and diet and host phylogeny are two of the most critical factors influencing the gut microbiome. A stable gut bacterial community plays a pivotal role in the host's physiological development and immune health. A phage is a virus that directly infects bacteria, and phages' close associations and interactions with bacteria are essential for maintaining the stability of the gut bacterial community and the entire microbial ecosystem. Here, we utilized 99 published metagenomic datasets from 38 mammalian species to investigate the relationship (diversity and composition) and potential interactions between gut bacterial and phage communities and the impact of diet and phylogeny on these communities. Our results highlight the co-evolutionary potential of bacterial-phage interactions within the mammalian gut. We observed a higher alpha diversity in gut bacteria than in phages and identified positive correlations between bacterial and phage compositions. Furthermore, our study revealed the significant influence of diet and phylogeny on mammalian gut bacterial and phage communities. We discovered that the impact of dietary factors on these communities was more pronounced than that of phylogenetic factors at the order level. In contrast, phylogenetic characteristics had a more substantial influence at the family level. The similar omnivorous dietary preference and closer phylogenetic relationship (family Ursidae) may contribute to the similarity of gut bacterial and phage communities between captive giant panda populations (GPCD and GPYA) and omnivorous animals (OC; including Sun bear, brown bear, and Asian black bear). This study employed co-occurrence microbial network analysis to reveal the potential interaction patterns between bacteria and phages. Compared to other mammalian groups (carnivores, herbivores, and omnivores), the gut bacterial and phage communities of bamboo-eating species (giant pandas and red pandas) exhibited a higher level of interaction. Additionally, keystone species and modular analysis showed the potential role of phages in driving and maintaining the interaction patterns between bacteria and phages in captive giant pandas. In sum, gaining a comprehensive understanding of the interaction between the gut microbiota and phages in mammals is of great significance, which is of great value in promoting healthy and sustainable mammals and may provide valuable insights into the conservation of wildlife populations, especially endangered animal species.
Collapse
Affiliation(s)
| | | | | | - Lifeng Zhu
- College of Life Sciences, Nanjing Normal University, Nanjing 210098, China; (M.Z.); (Y.Z.); (X.C.)
| |
Collapse
|
35
|
Olson M, Toffoli S, Vander Wyst KB, Zhou F, Reifsnider E, Petrov ME, Whisner CM. Associations of Infant Feeding, Sleep, and Weight Gain with the Toddler Gut Microbiome. Microorganisms 2024; 12:549. [PMID: 38543600 PMCID: PMC10972346 DOI: 10.3390/microorganisms12030549] [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: 01/12/2024] [Revised: 03/02/2024] [Accepted: 03/08/2024] [Indexed: 05/05/2024] Open
Abstract
This study examines how feeding, sleep, and growth during infancy impact the gut microbiome (GM) in toddlers. The research was conducted on toddlers (n = 36), born to Latina women of low-income with obesity. Their mothers completed retrospective feeding and sleeping questionnaires at 1, 6, and 12 months; at 36 months, fecal samples were collected. Sequencing of the 16S rRNA gene (V4 region) revealed that breastfeeding for at least 1 month and the introduction of solids before 6 months differentiated the GM in toddlerhood (Bray-Curtis, pseudo-F = 1.805, p = 0.018, and pseudo-F = 1.651, p = 0.044, respectively). Sleep had an effect across time; at 1 and 6 months of age, a lower proportion of nighttime sleep (relative to 24 h total sleep) was associated with a richer GM at three years of age (Shannon H = 4.395, p = 0.036 and OTU H = 5.559, p = 0.018, respectively). Toddlers experiencing rapid weight gain from birth to 6 months had lower phylogenetic diversity (Faith PD H = 3.633, p = 0.057). These findings suggest that early life nutrition, sleeping patterns, and growth rate in infancy may influence the GM composition. Further verification of these results with objective sleep data and a larger sample is needed.
Collapse
Affiliation(s)
- Magdalena Olson
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA; (M.O.); (S.T.); (K.B.V.W.); (F.Z.)
- Center for Health Through Microbiomes, The Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
| | - Samantha Toffoli
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA; (M.O.); (S.T.); (K.B.V.W.); (F.Z.)
| | - Kiley B. Vander Wyst
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA; (M.O.); (S.T.); (K.B.V.W.); (F.Z.)
| | - Fang Zhou
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA; (M.O.); (S.T.); (K.B.V.W.); (F.Z.)
| | - Elizabeth Reifsnider
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ 85004, USA; (E.R.); (M.E.P.)
| | - Megan E. Petrov
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ 85004, USA; (E.R.); (M.E.P.)
| | - Corrie M. Whisner
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA; (M.O.); (S.T.); (K.B.V.W.); (F.Z.)
- Center for Health Through Microbiomes, The Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
| |
Collapse
|
36
|
Rosen-Carole CB, Greenman S, Wang H, Sonawane S, Misra R, O'Connor T, Järvinen K, D'Angio C, Young BE. Association between maternal stress and premature milk cortisol, milk IgA, and infant health: a cohort study. Front Nutr 2024; 11:1270523. [PMID: 38533463 PMCID: PMC10964987 DOI: 10.3389/fnut.2024.1270523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 02/06/2024] [Indexed: 03/28/2024] Open
Abstract
Background Maternal stress is pervasive in the neonatal intensive care unit (NICU). Maternal stress is associated with changes in human milk (HM) immunomodulatory agents, which may impact neonatal health. We sought to determine the association between maternal stress, HM immunoglobulin A (IgA) and cortisol, and to assess how these milk components correlate with infant immune and neurodevelopmental outcomes. We then compared how these associations persist over time. Methods The study design involved a cohort study of exclusively breastfeeding mothers and their singleton moderately preterm (28-34 weeks) infants admitted to the NICU. We collected maternal serum, maternal saliva, and first-morning whole milk samples, and administered maternal stress questionnaires at 1 and 5 weeks postpartum. We analyzed the samples for HM IgA (using a customized immunoassay in skim milk) and for HM and salivary cortisol (using a chemiluminescent immunoassay). Infant illness was assessed using the Score for Neonatal Acute Physiology II (SNAP II) and SNAP II with Perinatal Extension (SNAPPE II), and infant neurodevelopment were assessed using the Test of Infant Motor Performance. We analyzed changes in HM IgA and cortisol over time using paired t-tests. Furthermore, we performed correlation and regression analyses after adjusting for gestational age (GA), corrected GA, and infant days of life. Results In our study, we enrolled 26 dyads, with a mean maternal age of 28.1 years, consisting of 69% white, 19% Black, and 8% Hispanic. Cortisol: Salivary and HM cortisol were closely associated in week 1 but not in week 5. Though mean salivary cortisol remained stable over time [2.41 ng/mL (SD 2.43) to 2.32 (SD 1.77), p = 0.17], mean HM cortisol increased [1.96 ng/mL (SD 1.93) to 5.93 ng/mL (SD 3.83), p < 0.001]. Stress measures were inversely associated with HM cortisol at week 1 but not at week 5. IgA: HM IgA decreased over time (mean = -0.14 mg/mL, SD 0.53, p < 0.0001). High maternal stress, as measured by the Parental Stressor Scale: neonatal intensive care unit (PSS:NICU), was positively associated with HM IgA at week 5 (r = 0.79, P ≤ 0.001). Higher IgA was associated with a lower (better) SNAP II score at week 1 (r = -0.74, p = 0.05). No associations were found between maternal stress, salivary cortisol, HM cortisol, or HM IgA and neurodevelopment at discharge (as assessed using the TIMP score). Furthermore, these relationships did not differ by infant sex. Conclusion Maternal stress showed associations with HM cortisol and HM IgA. In turn, HM IgA was associated with lower measures of infant illness.
Collapse
Affiliation(s)
- Casey B. Rosen-Carole
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Susan Greenman
- Swedish First Hill Family Medicine, Seattle, WA, United States
| | - Hongyue Wang
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Sharvari Sonawane
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Ravi Misra
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Tom O'Connor
- Department of Psychiatry, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Kirsi Järvinen
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Carl D'Angio
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Bridget E. Young
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| |
Collapse
|
37
|
Venter C, Meyer R, Bauer M, Bird JA, Fleischer DM, Nowak-Wegrzyn A, Anagnostou A, Vickery BP, Wang J, Groetch M. Identifying Children at Risk of Growth and Nutrient Deficiencies in the Food Allergy Clinic. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2024; 12:579-589. [PMID: 38280452 DOI: 10.1016/j.jaip.2024.01.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/16/2024] [Accepted: 01/19/2024] [Indexed: 01/29/2024]
Abstract
BACKGROUND Food allergies affect growth in children by decreasing the availability of nutrients through decreased dietary intake, increased dietary needs, food-medication interactions, and psychosocial burden. Guidelines on food allergy management frequently recommend nutrition counseling and growth monitoring of children with food allergies. OBJECTIVE To provide clear guidance for clinicians to identify children with food allergies who are at nutritional risk and ensure prompt intervention. METHODS We provide a narrative review summarizing information from national and international guidelines, retrospective studies, population studies, review articles, case reports, and case series to identify those with food allergy at greatest nutritional risk, determine the impact of nutritional interventions on growth, and develop guidance for risk reduction in children with food allergies. RESULTS Children with food allergies are at increased risk of nutritional deficiencies and poor growth. Nutritional assessment and intervention can improve outcomes. Identifying poor growth is an important step in the nutrition assessment. Therefore, growth should be assessed at each allergy evaluation. Interventions to ensure adequate dietary intake for growth include appropriately prescribed elimination diets, breast-feeding support and assessment, supplemental formula, vitamin and/or mineral supplementation, appropriate milk substitutes, and timely introduction of nutrient-dense complementary foods. Access to foods of appropriate nutritional value is an ongoing concern. CONCLUSION Nutrition intervention or referral to registered dietitian nutritionists with additional training and/or experience in food allergy may result in improved growth and nutrition outcomes.
Collapse
Affiliation(s)
- Carina Venter
- Section of Pediatric Allergy and Immunology, Children's Hospital Colorado, University of Colorado, Aurora, Colo.
| | - Rosan Meyer
- Department of Medicine, Imperial College London, London, United Kingdom; Department of Nutrition and Dietetics, University of Winchester, Winchester, United Kingdom; Department of Medicine, KU Leuven, Leuven, Belgium
| | - Maureen Bauer
- Section of Pediatric Allergy and Immunology, Children's Hospital Colorado, University of Colorado, Aurora, Colo
| | - J Andrew Bird
- Department of Pediatrics, Division of Allergy and Immunology, UT Southwestern Medical Center, Dallas, Texas
| | - David M Fleischer
- Section of Pediatric Allergy and Immunology, Children's Hospital Colorado, University of Colorado, Aurora, Colo
| | - Anna Nowak-Wegrzyn
- Hassenfeld Children's Hospital, Department of Pediatrics, NYU Grossman School of Medicine, New York, NY; Department of Pediatrics, Gastroenterology and Nutrition, Collegium Medicum, University of Warmia and Mazury, Olsztyn, Poland
| | - Aikaterini Anagnostou
- Section of Allergy and Immunology, Baylor College of Medicine, Houston, Texas; Section of Allergy and Immunology, Department of Pediatrics, Texas Children's Hospital, Houston, Texas
| | - Brian P Vickery
- Children's Healthcare of Atlanta, Atlanta, Ga; Department of Pediatrics, Emory University, Atlanta, Ga
| | - Julie Wang
- Division of Pediatric Allergy and Immunology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Marion Groetch
- Division of Pediatric Allergy and Immunology, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
38
|
Liu P, Liu Y, Cheng J, Xia Y, Yang Y. Copper exposure causes alteration in the intestinal microbiota and metabolites in Takifugu rubripes. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 272:116064. [PMID: 38340599 DOI: 10.1016/j.ecoenv.2024.116064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/22/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024]
Abstract
Copper is an environmental pollutant, and copper in aquatic environments mainly comes from soil and water. It enters the environment through atmospheric deposition, sewage discharge, and industrial production, and enters aquatic organisms, causing toxicity. Takifugu rubripes (T. rubripes) is a marine fish with high economic value. Due to the toxic effects of heavy metals on aquatic organisms such as fish, it can affect the gut community and metabolites of fish. The gut is an important channel for fish to communicate with the outside world and a necessary pathway for the metabolism of nutrients and toxic substances in the fish body. Studies have shown that due to changes in global water emissions and the high sensitivity of aquatic organisms to the environment, copper may pose greater potential hazards to aquatic organisms. Copper poses a greater risk to aquatic species than other heavy metals and metal/metal like pollutants (such as cadmium, lead, mercury, arsenic, etc.) . In order to elucidate the effects of copper exposure on the gut of T. rubripes. In this study, we exposed T. rubripes to 0, 50, 100, or 500 μg/L of copper for three days, the effects of copper exposure on the gut microbiota structure and metabolites of the T. rubripes were investigated using 16 S rRNA gene and metabolomics techniques. The research results indicate that with the increase copper concentration, the intestinal tissue of T. rubripes undergoes significant damage. 16 S rRNA sequencing results show that copper exposure alters the structure and metabolites of intestinal microbiota. Copper exposure of 100 and 500 μg/L inhibited the colonization of the bacterial gut, disrupted the intestinal barrier, and made the fish susceptible to the pathogens. Liquid chromatography-mass spectrometry analysis showed that copper regulated the production of metabolites such as L-histidine, arachidonic acid, and L-glutamic acid, which are related to energy and immunity. Microbiome-metabolome correlation analysis showed that Subdoligranulum, Family_XIII_AD3011_group, and Clostridium_sensu_stricto_1 were the key bacteria for copper ion intervention, and they might up-regulate the levels of metabolites such as indole-3-acetic acid, 3-indoleacrylic acid, and 5-hydroxyindole in the tryptophan metabolism pathway. In summary, our research has demonstrated that copper exposure can cause pathological changes in the intestinal tissue of the T. rubripes. High concentrations of copper ions can affect the colonization of the T. rubripes microbiota in the intestine, damage the fish's immune system, and alter the structure and metabolites of the intestinal microbiota, this can lead to intestinal metabolic dysfunction. providing a reference for the evaluation of the biological toxicity effects of heavy metal elements in the marine environment. This study provides a reference for evaluating the biological toxicity effects of heavy metal elements in marine environments.
Collapse
Affiliation(s)
- Pengfei Liu
- Key Laboratory of Environment Controlled Aquaculture (Dalian Ocean University) Ministry of Education, 116023, China; College of Marine Technology and Environment, Dalian Ocean University, Dalian 116023, China.
| | - Yanyun Liu
- Key Laboratory of Environment Controlled Aquaculture (Dalian Ocean University) Ministry of Education, 116023, China; College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China
| | - Jianxin Cheng
- Key Laboratory of Environment Controlled Aquaculture (Dalian Ocean University) Ministry of Education, 116023, China; College of Life Science, Liaoning Normal University, Dalian 116081, China
| | - Yuqing Xia
- Key Laboratory of Environment Controlled Aquaculture (Dalian Ocean University) Ministry of Education, 116023, China; School of Marine Sciences, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Yi Yang
- Key Laboratory of Environment Controlled Aquaculture (Dalian Ocean University) Ministry of Education, 116023, China; College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China
| |
Collapse
|
39
|
Koç N, Ersoy N, Yardimci H, Külhaş Çelik İ, Civelek E. Evaluation of Healthy Eating Index and Children's Diet Inflammatory Index according to asthma severity group. BMC Pediatr 2024; 24:127. [PMID: 38365643 PMCID: PMC10874057 DOI: 10.1186/s12887-023-04507-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 12/22/2023] [Indexed: 02/18/2024] Open
Abstract
BACKGROUND Diet may contribute to better asthma control in children by impacting the immune and inflammatory pathophysiology. Therefore, this study aimed to investigate differences in nutrient intake, Children's Dietary Inflammatory Index (C-DII), and dietary quality according to asthma severity. MATERIALS AND METHODS Asthma severity, dietary inflammatory status, and diet quality were assessed in a sample of 202 children with asthma (55.6% males, aged 5-18 years) attending a pediatric allergy outpatient clinic. Asthma severity was evaluated according to the Global Initiative for Asthma criteria and categorized as mild, moderate, or severe. The Children's Dietary Inflammatory Index (C-DII) and Healthy Eating Index (HEI-2010) were calculated based on information collected by the 24-h dietary recall method. Dietary quality was categorized as poor, moderate, or good diet according to HEI-2010. RESULTS The mean age of the participants was 9.6 ± 3.2 years. Children with severe asthma were younger on average (p < 0.05). Children with mild asthma had significantly higher fiber and iron intake than those with moderate asthma (p < 0.05). C-DII values did not differ significantly according to asthma severity (p > 0.05). Dietary quality was evaluated as moderate in 89.1% of the participants and also showed no difference based on asthma severity. CONCLUSIONS These findings suggest that inflammatory status and diet quality may not affect asthma severity in children, highlighting the influence of various genetic and environmental factors on the association between diet and asthma severity. More comprehensive and longitudinal studies are needed to investigate the mechanisms linking diet and asthma.
Collapse
Affiliation(s)
- Nevra Koç
- Department of Nutrition and Dietetics, Gülhane Health Sciences of Faculty, Sağlık Bilimleri University, İstanbul, Turkey.
| | - Nursena Ersoy
- Ankara University, Institute of Health Sciences, Ankara, Turkey
| | - Hülya Yardimci
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Ankara University, Ankara, Turkey
| | - İlknur Külhaş Çelik
- Division of Pediatric Allergy Immunology, Faculty of Medicine, Selcuk University, Konya, Turkey
| | - Ersoy Civelek
- Division of Pediatric Allergy Immunology, Sağlık Bilimleri University, Ankara City Hospital, Ankara, Turkey
| |
Collapse
|
40
|
Ruan Q, Jiang Y, Shi Y. Maternal smoking around birth and its influence on offspring allergic diseases: A mendelian randomization study. World Allergy Organ J 2024; 17:100875. [PMID: 38351904 PMCID: PMC10862070 DOI: 10.1016/j.waojou.2024.100875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 01/22/2024] [Indexed: 02/16/2024] Open
Abstract
Objective The influence of maternal smoking around birth (MSAB) on offspring allergic diseases, specifically childhood asthma (CA), allergic rhinitis (AR), allergic conjunctivitis (AC), and atopic dermatitis (AD) remains incompletely understood. We performed a rigorous mendelian randomization (MR) study to obtain the unconfounded association between MSAB and allergic diseases in offspring with and without adjustment for the effect of breastfeeding. Methods Utilizing publicly available information of MSAB, breastfeeding, CA, AR, AC, and AD from large-scale genome-wide association studies (GWAS), we performed a two-sample mendelian randomization (TSMR) analysis to assess the respective causal relationship of MSAB and breastfeeding to allergic diseases in offspring. To get a reliable conclusion, MR Egger regression, weighted median, and inverse variance weighted (IVW) were employed to estimate the causality, with IVW as the primary analysis. Multivariate MR (MVMR) analysis was used to assess the effect of MSAB on allergic diseases after adjusting for breastfeeding's impact. Sensitivity analysis was conducted using the Cochran Q test, MR-Egger, and leave-one-out approaches to ensure the reliability and stability of results. Results The TSMR analysis demonstrated MSAB increased the risks of CA (PIVW = 0.013, OR: 1.018, 95%CI: 1.004 to 1.033) and AD (PIVW = 0.006, OR: 8.293, 95%CI: 1.815 to 37.884) in offspring. Conversely, breastfeeding decreased the risk of CA (PIVW <0.001, OR: 0.946, 95%CI: 0.918 to 0.974). MSAB still increased the risks of CA (P = 0.0497, OR: 1.013, 95%CI: 1.000017 to 1.026) and AD (P = 0.003, OR: 13.800, 95%CI: 2.490 to 269.246) after adjusting for breastfeeding. We observed no strong indication of a negative causality between MSAB and AC and AR. Conclusion Our findings provided robust evidence of the adverse effects of MSAB on offspring. We emphasized the urgency of smoking cessation around birth and the importance of breastfeeding even in smoking mothers.
Collapse
Affiliation(s)
- Qiqi Ruan
- Department of Neonatology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, People's Republic of China
| | - Yu Jiang
- Department of Pediatrics, Women and Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Yuan Shi
- Department of Neonatology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, People's Republic of China
| |
Collapse
|
41
|
Liang C, Niu HY, Lyu LZ, Wu YF, Zhang LW. Profiles of Intestinal Flora in Breastfed Obese Children and Selecting Functional Strains Against Obesity. Mol Nutr Food Res 2024; 68:e2300735. [PMID: 38227364 DOI: 10.1002/mnfr.202300735] [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: 10/16/2023] [Revised: 11/22/2023] [Indexed: 01/17/2024]
Abstract
SCOPE Breast milk has the potential to prevent childhood obesity by providing probiotics, but there are still instances of obesity in breastfed children. METHODS AND RESULTS This study investigates the difference in intestinal flora structure between breastfed children with obesity (OB-BF) and normal-weight breastfed children (N-BF). Building upon this foundation, it employs both cell and mouse models to identify an antiobesity strain within the fecal matter of N-BF children and explore its underlying mechanisms. The results reveal a reduction in lactobacillus levels within the intestinal flora of OB-BF children compared to N-BF children. Consequently, Lactobacillus plantarum H-72 (H-72) is identified as a promising candidate due to its capacity to stimulate glucagon-like peptide-1 (GLP-1) secretion in enteroendocrine cells (ECCs). In vivo, H-72 effectively increases serum GLP-1 concentration, reduces food intake, regulates the expression of genes related to energy metabolism (SCD-1, FAS, UCP-1, and UCP-3), and regulates gut microbiota structure in mice. Moreover, the lipoteichoic acid of H-72 activates toll-like receptor 4 to enhanced GLP-1 secretion in STC-1 cells. CONCLUSIONS L. plantarum H-72 is screened out for its potential antiobesity effect, which presents a potential and promising avenue for future interventions aimed at preventing pediatric obesity in breastfed children.
Collapse
Affiliation(s)
- Cong Liang
- College of Safety and Environmental Engineering, Shandong University of Science and Technology, Qingdao, 266510, China
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150010, China
| | - Hai-Yue Niu
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150010, China
| | - Lin-Zheng Lyu
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150010, China
| | - Yi-Fan Wu
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150010, China
| | - Lan-Wei Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, China
| |
Collapse
|
42
|
Catassi G, Aloi M, Giorgio V, Gasbarrini A, Cammarota G, Ianiro G. The Role of Diet and Nutritional Interventions for the Infant Gut Microbiome. Nutrients 2024; 16:400. [PMID: 38337684 PMCID: PMC10857663 DOI: 10.3390/nu16030400] [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: 12/14/2023] [Revised: 01/14/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
The infant gut microbiome plays a key role in the healthy development of the human organism and appears to be influenced by dietary practices through multiple pathways. First, maternal diet during pregnancy and infant nutrition significantly influence the infant gut microbiota. Moreover, breastfeeding fosters the proliferation of beneficial bacteria, while formula feeding increases microbial diversity. The timing of introducing solid foods also influences gut microbiota composition. In preterm infants the gut microbiota development is influenced by multiple factors, including the time since birth and the intake of breast milk, and interventions such as probiotics and prebiotics supplementation show promising results in reducing morbidity and mortality in this population. These findings underscore the need for future research to understand the long-term health impacts of these interventions and for further strategies to enrich the gut microbiome of formula-fed and preterm infants.
Collapse
Affiliation(s)
- Giulia Catassi
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (G.C.); (A.G.); (G.C.)
- Pediatric Gastroenterology and Liver Unit, Sapienza University of Rome, Umberto I Hospital, 00161 Rome, Italy;
| | - Marina Aloi
- Pediatric Gastroenterology and Liver Unit, Sapienza University of Rome, Umberto I Hospital, 00161 Rome, Italy;
| | - Valentina Giorgio
- Department of Woman and Child Health and Public Health, UOC Pediatria, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy;
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (G.C.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Giovanni Cammarota
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (G.C.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Gianluca Ianiro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (G.C.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|
43
|
Biagioli V, Volpedo G, Riva A, Mainardi P, Striano P. From Birth to Weaning: A Window of Opportunity for Microbiota. Nutrients 2024; 16:272. [PMID: 38257165 PMCID: PMC10819289 DOI: 10.3390/nu16020272] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/12/2024] [Accepted: 01/14/2024] [Indexed: 01/24/2024] Open
Abstract
(1) Background: The first 1000 days of life constitute a critical window of opportunity for microbiota development. Nutrients play a crucial role in enriching and diversifying the microbiota, derived not only from solid food but also from maternal dietary patterns during gestation. (2) Methods: We conducted a comprehensive literature review using the PubMed database, covering eleven years (2013-2023). We included English-language reviews, original research papers, and meta-analyses, while excluding case reports and letters. (3) Results: Consensus in the literature emphasizes that our interaction with a multitude of microorganisms begins in the intrauterine environment and continues throughout our lives. The existing data suggest that early nutritional education programs, initiated during pregnancy and guiding infant diets during development, may influence the shaping of the gut microbiota, promoting long-term health. (4) Conclusions: Further research is necessary in the coming years to assess potential interventions and early nutritional models aimed at modulating the pediatric microbiota, especially in vulnerable populations such as premature newborns.
Collapse
Affiliation(s)
- Valentina Biagioli
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16126 Genoa, Italy; (G.V.); (A.R.); (P.M.); (P.S.)
| | - Greta Volpedo
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16126 Genoa, Italy; (G.V.); (A.R.); (P.M.); (P.S.)
| | - Antonella Riva
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16126 Genoa, Italy; (G.V.); (A.R.); (P.M.); (P.S.)
| | - Paolo Mainardi
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16126 Genoa, Italy; (G.V.); (A.R.); (P.M.); (P.S.)
| | - Pasquale Striano
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16126 Genoa, Italy; (G.V.); (A.R.); (P.M.); (P.S.)
- IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| |
Collapse
|
44
|
Nuzum ND, Deady C, Kittel-Schneider S, Cryan JF, O'Mahony SM, Clarke G. More than just a number: the gut microbiota and brain function across the extremes of life. Gut Microbes 2024; 16:2418988. [PMID: 39567371 PMCID: PMC11583591 DOI: 10.1080/19490976.2024.2418988] [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: 05/28/2024] [Revised: 09/24/2024] [Accepted: 10/14/2024] [Indexed: 11/22/2024] Open
Abstract
Understanding the interrelationship between the gut microbiota and host physiology, although still in its relative infancy, has taken important steps forward over the past decade. In the context of brain disorders including those characterized by neurodevelopmental and neurodegenerative changes there have been important advances. However, initially research involved correlational analyses, had limited translational scope, and lacked functional assessments. Thus, largescale longitudinal clinical investigations that assess causation and underlying mechanisms via in depth analysis methods are needed. In neurodegeneration research, strong causal evidence now links the gut microbiome to Alzheimer's (AD), and Parkinson's Disease (PD), as supported by human-to-animal transplantation studies. Longitudinal interventions are being conducted in AD, PD, amyotrophic lateral sclerosis, Huntington's disease, and multiple sclerosis. Neurodevelopmental research has also seen a boon in microbiome-related clinical research including in autism, Attention-deficit/hyperactivity disorder, and schizophrenia, which is confirming prior animal model work regarding the key time-windows in the gut microbiome important for infant cognition. While recent research advances represent important progress, fundamental knowledge gaps and obstacles remain. Knowing how and why the gut microbiome changes at the extremes of life will develop our mechanistic understanding and help build the evidence base as we strive toward counteracting microbial missteps with precision therapeutic interventions.
Collapse
Affiliation(s)
- Nathan D Nuzum
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Clara Deady
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Sarah Kittel-Schneider
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Siobhain M O'Mahony
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| |
Collapse
|
45
|
Yang J, Chen X, Liu T, Shi Y. Potential role of bile acids in the pathogenesis of necrotizing enterocolitis. Life Sci 2024; 336:122279. [PMID: 37995935 DOI: 10.1016/j.lfs.2023.122279] [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: 09/01/2023] [Revised: 11/13/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023]
Abstract
Necrotizing enterocolitis (NEC) is one of the most common acute gastrointestinal diseases in preterm infants. Recent studies have found that NEC is not only caused by changes in the intestinal environment but also by the failure of multiple systems and organs, including the liver. The accumulation of bile acids (BAs) in the ileum and the disorder of ileal BA transporters are related to the ileum injury of NEC. Inflammatory factors such as tumor necrosis factor (TNF)-α and interleukin (IL)-18 secreted by NEC also play an important role in regulating intrahepatic BA transporters. As an important link connecting the liver and intestinal circulation, the bile acid metabolic pathway plays an important role in the regulation of intestinal microbiota, cell proliferation, and barrier protection. In this review, we focus on how bile acids explore the dynamic changes of bile acid metabolism in necrotizing enterocolitis and the potential therapeutic value of targeting the bile acid signaling pathways.
Collapse
Affiliation(s)
- Jiahui Yang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Xiaoyu Chen
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Tianjing Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Yongyan Shi
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
46
|
Giannì ML, Morniroli D, Mosca F, Rescigno M. Can Postbiotics Represent a New Strategy for NEC? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1449:43-57. [PMID: 39060730 DOI: 10.1007/978-3-031-58572-2_3] [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: 07/28/2024]
Abstract
Intestinal bacteria, also known as gut microbiota, are a rich ecology of microorganisms found in the human digestive tract. Extensive study has highlighted their critical relevance in preserving human health. New research has revealed that bacterial viability is not invariably necessary to induce health benefits. Postbiotics (defined soluble substances produced as a byproduct of the metabolic processes of living microbes) have thus emerged as an important topic of research. They contribute to shaping the gut microbiota, exert immune-modulation activity, and improve the integrity of the gut barrier.Alterations in preterm gut colonization associated with intestinal barrier immaturity and the increased reactivity of the intestinal mucosa to colonizing bacteria have been implicated in the pathogenesis of necrotizing enterocolitis. Postbiotics have shown promising outcomes in reducing the risk of developing NEC, lowering inflammation, encouraging the development of good bacteria, and strengthening the intestinal barrier. This is an important advancement in newborn care and highlights the potential of postbiotics to avoid severe intestinal disorders.
Collapse
Affiliation(s)
- Maria Lorella Giannì
- Fondazione I.R.C.C.S. Ca' Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit, Milan, Italy
- Department of Clinical Science and Community Health, University of Milan, Milan, Italy
| | - Daniela Morniroli
- Fondazione I.R.C.C.S. Ca' Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit, Milan, Italy
- Department of Clinical Science and Community Health, University of Milan, Milan, Italy
| | - Fabio Mosca
- Fondazione I.R.C.C.S. Ca' Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit, Milan, Italy.
- Department of Clinical Science and Community Health, University of Milan, Milan, Italy.
| | | |
Collapse
|
47
|
Zuurveld M, Diks MAP, Kiliaan PCJ, Garssen J, Folkerts G, van’t Land B, Willemsen LEM. Butyrate interacts with the effects of 2'FL and 3FL to modulate in vitro ovalbumin-induced immune activation, and 2'FL lowers mucosal mast cell activation in a preclinical model for hen's egg allergy. Front Nutr 2023; 10:1305833. [PMID: 38174112 PMCID: PMC10762782 DOI: 10.3389/fnut.2023.1305833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 11/15/2023] [Indexed: 01/05/2024] Open
Abstract
Background Early life provides a window of opportunity to prevent allergic diseases. With a prevalence of 0.5-2% in infants, hen's egg allergy is one of the most common food allergies. The immunomodulatory effects of human milk oligosaccharides (HMOs), 2'-fucosyllactose (2'FL), and 3-fucosyllactose (3FL) were studied in an in vitro mucosal immune model and an in vivo murine model for hen's egg (ovalbumin) allergy. Methods Intestinal epithelial cell (IEC)/dendritic cell (DC) and DC/T cell cocultures were used to expose IECs to ovalbumin (OVA) in an in vitro mucosal immune model. The effects of epithelial pre-incubation with 0.1% 2'FL or 3FL and/or 0.5 mM butyrate were studied. Three- to four-weeks-old female C3H/HeOuJ mice were fed AIN93G diets containing 0.1-0.5% 2'FL or 3FL 2 weeks before and during OVA sensitization and challenge. Allergic symptoms and systemic and local immune parameters were assessed. Results Exposing IECs to butyrate in vitro left the IEC/DC/T cell cross-talk unaffected, while 2'FL and 3FL showed differential immunomodulatory effects. In 3FL exposed IEC-DC-T cells, the secretion of IFNγ and IL10 was enhanced. This was observed upon pre-incubation of IECs with 2'FL and butyrate as well, but not 2'FL alone. The presence of butyrate did not affect OVA activation, but when combined with 3FL, an increase in IL6 release from DCs was observed (p < 0.001). OVA allergic mice receiving 0.5% 3FL diet had a lower %Th2 cells in MLNs, but the humoral response was unaltered compared to control mice. OVA-allergic mice receiving 0.1 or 0.5% 2'FL diets had lower serum levels of OVA-IgG2a (p < 0.05) or the mast cell marker mMCP1, in association with increased concentration of cecal short-chain fatty acids (SCFAs) (p < 0.05). Conclusion In vitro butyrate exposure promotes the development of a downstream type 1 and regulatory response observed after 2'FL exposure. 2'FL and 3FL differentially modulate ovalbumin-induced mucosal inflammation predominantly independent of butyrate. Mice receiving dietary 3FL during ovalbumin sensitization and challenge had lowered Th2 activation while the frequency of Treg cells was enhanced. By contrast, 2'FL improved the humoral immune response and suppressed mast cell activation in association with increased SCFAs production in the murine model for hen's egg allergy.
Collapse
Affiliation(s)
- M. Zuurveld
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - M. A. P. Diks
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - P. C. J. Kiliaan
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - J. Garssen
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
- Danone Nutricia Research B.V, Utrecht, Netherlands
| | - G. Folkerts
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - B. van’t Land
- Danone Nutricia Research B.V, Utrecht, Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - L. E. M. Willemsen
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
48
|
Liu Y, Huang Y, He Q, Dou Z, Zeng M, Wang X, Li S. From heart to gut: Exploring the gut microbiome in congenital heart disease. IMETA 2023; 2:e144. [PMID: 38868221 PMCID: PMC10989834 DOI: 10.1002/imt2.144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/14/2023] [Accepted: 09/26/2023] [Indexed: 06/14/2024]
Abstract
Congenital heart disease (CHD) is a prevalent birth defect and a significant contributor to childhood mortality. The major characteristics of CHD include cardiovascular malformations and hemodynamical disorders. However, the impact of CHD extends beyond the circulatory system. Evidence has identified dysbiosis of the gut microbiome in patients with CHD. Chronic hypoxia and inflammation associated with CHD affect the gut microbiome, leading to alterations in its number, abundance, and composition. The gut microbiome, aside from providing essential nutrients, engages in direct interactions with the host immune system and indirect interactions via metabolites. The abnormal gut microbiome or its products can translocate into the bloodstream through an impaired gut barrier, leading to an inflammatory state. Metabolites of the gut microbiome, such as short-chain fatty acids and trimethylamine N-oxide, also play important roles in the development, treatment, and prognosis of CHD. This review discusses the role of the gut microbiome in immunity, gut barrier, neurodevelopment, and perioperative period in CHD. By fostering a better understanding of the cross-talk between CHD and the gut microbiome, this review aims to contribute to improve clinical management and outcomes for CHD patients.
Collapse
Affiliation(s)
- Yuze Liu
- Pediatric Cardiac Surgery Centre, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Yuan Huang
- Pediatric Cardiac Surgery Centre, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Qiyu He
- Pediatric Cardiac Surgery Centre, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Zheng Dou
- Pediatric Cardiac Surgery Centre, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Min Zeng
- Department of Pediatric Intensive Care Unit, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Xu Wang
- Department of Pediatric Intensive Care Unit, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Shoujun Li
- Pediatric Cardiac Surgery Centre, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| |
Collapse
|
49
|
Grant ET, Boudaud M, Muller A, Macpherson AJ, Desai MS. Maternal diet and gut microbiome composition modulate early-life immune development. EMBO Mol Med 2023; 15:e17241. [PMID: 37278126 PMCID: PMC10405054 DOI: 10.15252/emmm.202217241] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 06/07/2023] Open
Abstract
In early life, the intestinal mucosa and immune system undergo a critical developmental process to contain the expanding gut microbiome while promoting tolerance toward commensals, yet the influence of maternal diet and microbial composition on offspring immune maturation remains poorly understood. We colonized germ-free mice with a consortium of 14 strains, fed them a standard fiber-rich chow or a fiber-free diet, and then longitudinally assessed offspring development during the weaning period. Unlike pups born to dams fed the fiber-rich diet, pups of fiber-deprived dams demonstrated delayed colonization with Akkermansia muciniphila, a mucin-foraging bacterium that can also use milk oligosaccharides. The pups of fiber-deprived dams exhibited an enrichment of colonic transcripts corresponding to defense response pathways and a peak in Il22 expression at weaning. Removal of A. muciniphila from the community, but maintenance on the fiber-rich diet, was associated with reduced proportions of RORγt-positive innate and adaptive immune cell subsets. Our results highlight the potent influence of maternal dietary fiber intake and discrete changes in microbial composition on the postnatal microbiome assemblage and early immune development.
Collapse
Affiliation(s)
- Erica T Grant
- Department of Infection and ImmunityLuxembourg Institute of HealthEsch‐sur‐AlzetteLuxembourg
- Faculty of Science, Technology and MedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Marie Boudaud
- Department of Infection and ImmunityLuxembourg Institute of HealthEsch‐sur‐AlzetteLuxembourg
| | - Arnaud Muller
- Bioinformatics Platform, Data Integration and AnalysisLuxembourg Institute of HealthStrassenLuxembourg
- LuxGen, Translational Medicine Operation HubLuxembourg Institute of HealthDudelangeLuxembourg
| | - Andrew J Macpherson
- Department of Visceral Surgery and MedicineBern University Hospital, University of BernBernSwitzerland
| | - Mahesh S Desai
- Department of Infection and ImmunityLuxembourg Institute of HealthEsch‐sur‐AlzetteLuxembourg
| |
Collapse
|
50
|
Taddei CR, Neu J. Editorial: Microbiome in the first 1000 days: multi-omic interactions, physiological effects, and clinical implications. Front Cell Infect Microbiol 2023; 13:1242626. [PMID: 37457954 PMCID: PMC10348902 DOI: 10.3389/fcimb.2023.1242626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/18/2023] Open
Affiliation(s)
- Carla R. Taddei
- School of Arts, Sciences and Humanity, University of São Paulo, São Paulo, Brazil
- Division of Clinical Laboratory, University Hospital - University of São Paulo, São Paulo, Brazil
| | - Josef Neu
- Division of Neonatology, University of Florida College of Medicine, Gainesville, FL, United States
| |
Collapse
|