1
|
Zhang D, Xie D, Qu Y, Mu D, Wang S. Digging deeper into necrotizing enterocolitis: bridging clinical, microbial, and molecular perspectives. Gut Microbes 2025; 17:2451071. [PMID: 39826099 DOI: 10.1080/19490976.2025.2451071] [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: 10/04/2024] [Revised: 12/26/2024] [Accepted: 01/02/2025] [Indexed: 01/20/2025] Open
Abstract
Necrotizing Enterocolitis (NEC) is a severe, life-threatening inflammatory condition of the gastrointestinal tract, especially affecting preterm infants. This review consolidates evidence from various biomedical disciplines to elucidate the complex pathogenesis of NEC, integrating insights from clinical, microbial, and molecular perspectives. It emphasizes the modulation of NEC-associated inflammatory pathways by probiotics and novel biologics, highlighting their therapeutic potential. We further critically examine dysbiotic alterations within the gut microbiota, with a particular focus on imbalances in bacterial and viral communities, which may contribute to the onset of NEC. The intricate interactions among toll-like receptor 4 (TLR4), microvascular integrity, immune activation, and the inflammatory milieu are meticulously summarized, offering a sophisticated understanding of NEC pathophysiology. This academic review aims to enhance the etiological comprehension of NEC, promote the development of targeted therapeutic interventions, and impart the significant impact of perinatal factors on the formulation of preventive and curative strategies for the disease.
Collapse
Affiliation(s)
- Deshuang Zhang
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
- Division of Neonatology/Pediatric Surgery, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Dongke Xie
- Division of Neonatology/Pediatric Surgery, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yi Qu
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
| | - Dezhi Mu
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
| | - Shaopu Wang
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
2
|
Hashimoto R, Nishiyama K, Namai F, Suzuki K, Sakuma T, Fukuda I, Sugiyama Y, Okano K, Shanoh T, Toyoshi E, Ohgi R, Saha S, Tsuchida S, Nishiyama E, Mukai T, Furukawa M, Nochi T, Villena J, Ikeda-Ohtsubo W, Yoshioka G, Nakazaki E, Suda Y, Kitazawa H. Milk sialyl-oligosaccharides mediate the early colonization of gut commensal microbes in piglets. MICROBIOME 2025; 13:135. [PMID: 40413516 PMCID: PMC12103040 DOI: 10.1186/s40168-025-02129-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 04/30/2025] [Indexed: 05/27/2025]
Abstract
BACKGROUND The suckling period in pigs is a key phase in development for shaping the gut microbiota, which is essential for maintaining biological homeostasis in neonates. In piglets fed sow milk, the gut microbiota comprises predominantly lactobacilli, indicating a host-gut microbiota symbiosis that is influenced by sow milk components. In this study, we sought to elucidate the mechanisms underlying the establishment and maintenance of the gut microbiome in suckling piglets, with a specific focus on the metabolism of sialyl-oligosaccharides by lactobacilli. RESULTS Based on liquid chromatography-mass spectrometry analysis, we identified 3'-sialyl-lactose (3'SL) as the major oligosaccharide in porcine milk, and microbiome profiling revealed the predominance of Ligilactobacillus salivarius during the suckling period, with a subsequent transition to Limosilactobacillus reuteri dominance post-weaning. Notably, sialic acid metabolism was established to be exclusively attributable to L. salivarius, thereby highlighting the pivotal role of 3'SL in determining species-specific bacterial segregation. L. salivarius was found to metabolize 3'SL when co-cultured with Bacteroides thetaiotaomicron, resulting in a shift in the predominant short-chain fatty acid produced, from lactate to acetate. This metabolic shift, in turn, inhibits the growth of enterotoxigenic Escherichia coli. Furthermore, the comparison of the gut microbiota between suckling piglets and those fed a low-3'SL formula revealed distinct diversity profiles. We accordingly speculate that an absence of sialyl-oligosaccharides in the formula-fed piglets may have restricted the growth of sialic acid-utilizing bacteria such as L. salivarius, thereby leading to a higher abundance of Enterobacteriaceae. CONCLUSIONS Our findings reveal the influence of sialyl-oligosaccharides in promoting microbial diversity and gut homeostasis, thereby highlighting the importance of sialic acid as a key factor in shaping milk-driven microbial colonization during the early stages of piglet development. Video Abstract.
Collapse
Affiliation(s)
- Ryoga Hashimoto
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, 980-8572, Japan
| | - Keita Nishiyama
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, 980-8572, Japan.
- Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.
| | - Fu Namai
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, 980-8572, Japan
- Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Kasumi Suzuki
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, 980-8572, Japan
- Swine and Poultry Research Department, Gifu Prefectural Livestock Research Institute, Seki, Japan
| | - Taiga Sakuma
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, 980-8572, Japan
| | - Itsuko Fukuda
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Nada-Ku, Kobe, Hyogo, Japan
| | - Yuta Sugiyama
- Center for Food Science and Wellness, Gunma University, Gunma University, Maebashi, Japan
| | - Kenji Okano
- Department of Life Science & Biotechnology, Kansai University, Suita, Osaka, Japan
| | | | - Eita Toyoshi
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, 980-8572, Japan
| | - Ryusuke Ohgi
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, 980-8572, Japan
| | - Sudeb Saha
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, 980-8572, Japan
| | - Sae Tsuchida
- Biotechnological Research Support Division, FASMAC Co., Ltd, Atsugi, Japan
| | - Eri Nishiyama
- Biotechnological Research Support Division, FASMAC Co., Ltd, Atsugi, Japan
| | - Takao Mukai
- Department of Animal Science, School of Veterinary Medicine, Kitasato University, Aomori, Japan
| | - Mutsumi Furukawa
- Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Laboratory of Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, Japan
| | - Tomonori Nochi
- Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Laboratory of Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, Japan
| | - Julio Villena
- Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), 4000, Tucuman, Argentina
| | - Wakako Ikeda-Ohtsubo
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, 980-8572, Japan
- Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Gou Yoshioka
- Swine and Poultry Research Department, Gifu Prefectural Livestock Research Institute, Seki, Japan
| | | | - Yoshihito Suda
- Department of Food, Agriculture and Environment, Miyagi University, Sendai, Japan
| | - Haruki Kitazawa
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, 980-8572, Japan.
- Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.
| |
Collapse
|
3
|
Łyko M, Maj J, Jankowska-Konsur A. The Role of the Gut Microbiome in Non-Hodgkin Lymphoma (NHL): A Focus on Diffuse Large B-Cell Lymphoma, Follicular Lymphoma, Cutaneous T-Cell Lymphoma, and NK/T-Cell Lymphoma. Cancers (Basel) 2025; 17:1709. [PMID: 40427206 PMCID: PMC12110234 DOI: 10.3390/cancers17101709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Revised: 05/14/2025] [Accepted: 05/17/2025] [Indexed: 05/29/2025] Open
Abstract
Non-Hodgkin lymphomas (NHLs) encompass a diverse group of neoplasms arising from the clonal proliferation of B-cell progenitors, T-cell progenitors, mature B-cells, mature T-cells, and natural killer (NK) cells. These malignancies account for over 90% of lymphoid neoplasms. The link between the gut microbiome and neoplasms has been extensively studied in recent years. Growing evidence suggests that the gut microbiome may be involved not only in the development of the disease, but also in modulating the efficacy of implemented therapies. In this review, we summarize the current knowledge on the potential involvement of the gut microbiome in the development of diffuse large B-cell lymphoma (DLBCL), follicular lymphoma (FL), mucosa-associated lymphoid tissue (MALT) lymphoma, and NK/T-cell lymphoma, including cutaneous T-cell lymphoma (CTCL). Moreover, we discuss the relationship between gut microbiome changes before and after treatment and their association with treatment outcomes, focusing on chemotherapy and CAR T-cell therapy.
Collapse
Affiliation(s)
- Magdalena Łyko
- Clinical Department of Oncodermatology, University Centre of General Dermatology and Oncodermatology, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| | - Joanna Maj
- Clinical Department of General Dermatology, University Centre of General Dermatology and Oncodermatology, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| | - Alina Jankowska-Konsur
- Clinical Department of Oncodermatology, University Centre of General Dermatology and Oncodermatology, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| |
Collapse
|
4
|
Bosco A, Arru F, Abis A, Fanos V, Dessì A. Application of Multiomics in Perinatology: A Metabolomics Integration-Focused Review. Int J Mol Sci 2025; 26:4164. [PMID: 40362403 PMCID: PMC12071719 DOI: 10.3390/ijms26094164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 03/13/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Precision medicine stems from a new approach to the prevention, diagnosis and treatment of patients, due to the shift in focus away from pathology and towards the uniqueness of the individual, personalising the diagnostic-therapeutic pathway. This paradigm shift has been made possible by the emergence of new high-throughput technologies capable of generating large amounts of data on multiple levels of a biological system, identifying pathology-related genes, transcripts, proteins and metabolites. Metabolomics plays a primary role in this context, providing, through non-invasive sampling, a very close image of the phenotype of the organism being studied by detecting metabolites, end products downstream of gene transcription, present in cells, tissues, organs and biological fluids. The enormous amount of data that these modern technologies make available, together with the need to elucidate the complex interplay of the various biological levels by combining data from distinct omics, has led to the need to employ advanced informatics techniques, among which artificial intelligence has recently emerged. These innovations are of great interest in the field of perinatology, representing an attempt to optimise the diagnostic timeline for the most critical newborns. In addition, they may contribute to the improvement of prevention strategies available to date. All these contributions prove to be crucial at very vulnerable life stages, allowing crucial intervention opportunities. In this review, we have analysed studies that have integrated metabolomics with at least one other omics in the perinatal field, attempting to highlight the usefulness of multiomics integration and the different methods employed.
Collapse
Affiliation(s)
| | | | | | | | - Angelica Dessì
- Neonatal Intensive Care Unit, Department of Surgical Sciences, University of Cagliari, AOU Cagliari, 09124 Cagliari, Italy; (A.B.); (F.A.); (A.A.); (V.F.)
| |
Collapse
|
5
|
Thatrimontrichai A, Praditaukrit M, Maneenil G, Dissaneevate S, Singkhamanan K, Surachat K. Butyrate Producers in Very Low Birth Weight Infants with Neither Culture-Proven Sepsis nor Necrotizing Enterocolitis. Nutrients 2025; 17:1329. [PMID: 40284194 PMCID: PMC12029688 DOI: 10.3390/nu17081329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 04/04/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Severe infection (sInfection; either late-onset culture-proven sepsis or necrotizing enterocolitis) in very low birth weight (VLBW) infants increases mortality rates and may show long-term progression. The fecal microbiome composition in VLBW infants with and without sInfection was classified in the sInfection and non-sInfection groups. Methods: Gut microbiomes, secondary information from a previous randomized trial, were analyzed using QIIME 2 software. The biodiversity and abundance of the gut microbiota between the sInfection and non-sInfection groups were compared. Results: Fifty-one neonates were included in the sInfection (n = 9) and non-sInfection (n = 42) groups; no significant differences in the fecal microbiome were observed in both alpha and beta diversities. Analysis of relative abundance revealed that in both groups, the predominant gut microbiota phylum, class, and genus were Proteobacteria, Gammaproteobacteria, and Klebsiella, respectively. The main fecal microbiome in the non-sInfection group included Faecalibacterium, Clostridium butyricum, and Bacteroides fragilis. Clostridium_sensu_stricto _1 was significantly more abundant in the non-sInfection group than in the sInfection group. Conclusions: Clostridium_sensu_stricto_1 was the main gut microbiota in the non-sInfection group. Considering the potential taxa as synbiotics (correlations among prebiotics, probiotics, and postbiotics), therapeutics may be useful for preventing and managing necrotizing enterocolitis or late-onset culture-proven sepsis in VLBW infants.
Collapse
Affiliation(s)
- Anucha Thatrimontrichai
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (M.P.); (G.M.); (S.D.)
| | - Manapat Praditaukrit
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (M.P.); (G.M.); (S.D.)
| | - Gunlawadee Maneenil
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (M.P.); (G.M.); (S.D.)
| | - Supaporn Dissaneevate
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (M.P.); (G.M.); (S.D.)
| | - Kamonnut Singkhamanan
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (K.S.); (K.S.)
| | - Komwit Surachat
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (K.S.); (K.S.)
| |
Collapse
|
6
|
Yu L, Li Y, Zuming Y. Enhanced predictive accuracy of mortality in VLBW infants with late-onset sepsis through a time-specific nomogram. Front Public Health 2025; 13:1548695. [PMID: 40241944 PMCID: PMC11999929 DOI: 10.3389/fpubh.2025.1548695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
Objective This study aims to develop and validate a nomogram-based scoring system to predict mortality in very low birth weight (VLBW) infants with late-onset sepsis (LOS). Timely risk stratification in this vulnerable population is critical for optimizing clinical outcomes. Methods We conducted a retrospective analysis on 202 VLBW infants diagnosed with LOS between January 2018 and December 2022. Predictive models were created at three key time points: 0 h, 6 h, and 12 h post-sepsis onset, utilizing Least Absolute Shrinkage and Selection Operator (LASSO) regression for variable selection and multivariable logistic regression for model construction. Internal validation was performed with 1,000 bootstrap resamples to correct for potential overfitting. External validation was conducted on an independent cohort of 71 infants from January 2023 to March 2024. Model performance was assessed using Area Under the Curve (AUC), calibration plots, and decision curve analysis (DCA). Results The models exhibited excellent discrimination with AUCs of 0.83, 0.92, and 0.94 at 0 h, 6 h, and 12 h, respectively, in the development cohort, and 0.95, 0.95, and 0.97 in the validation cohort. Calibration plots showed strong agreement between predicted and observed outcomes. The significant disparity in maternal COVID-19 infection rates between cohorts (1 vs. 89%) may have contributed to the enhanced predictive accuracy in the external cohort. Conclusion This dynamic, time-specific nomogram demonstrates high predictive accuracy and clinical utility for mortality in VLBW infants with LOS. The impact of maternal COVID-19 infection on neonatal outcomes offers a novel perspective for future research in sepsis prognostication.
Collapse
Affiliation(s)
- Lun Yu
- Suzhou Municipal Hospital, Suzhou, China
| | - Yanhong Li
- Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | | |
Collapse
|
7
|
Toubon G, Patin C, Delannoy J, Rozé JC, Barbut F, Ancel PY, Charles MA, Butel MJ, Lepage P, Aires J. Very preterm gut microbiota development from the first week of life to 3.5 years of age: a prospective longitudinal multicenter study. Microbiol Spectr 2025; 13:e0163624. [PMID: 39969235 PMCID: PMC11960047 DOI: 10.1128/spectrum.01636-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 01/18/2025] [Indexed: 02/20/2025] Open
Abstract
This study analyzed the longitudinal evolution of intestinal microbiota in very preterm neonates (PN) during and after their hospitalization. The bacterial 16S rRNA gene sequencing approach was applied for the analysis of fecal samples (n = 1,307) from 596 PN. Samples were collected at one week after birth, at one month, at the neonatal intensive care unit discharge, and at 3.5 years of age. Over time, the intestinal microbiota of the infants matured progressively, with increasing alpha diversity and decreasing beta diversity. Based on a Dirichlet multinomial mixture clustering approach (DMM), during hospitalization, infants progressed among ten different clusters. At 3.5 years of age, only three clusters were identified. The influence of the gestational age, the neonatal antibiotic administration, and the maternal antibiotic therapy during delivery on the gut microbiota varied over time and depended on the sampling period. Preconceptional maternal body mass index (BMI) was associated with the gut microbiota of infants during the hospitalization period and at 3.5 years of age. Infants with a lower gestational age or those born by Cesarean section shifted between clusters more frequently. Using PICRUSt2, the inferred metabolic pathways revealed a change in the functional capacities of the intestinal microbiota over time. We found that preconceptional maternal BMI was the only consistent perinatal factor influencing the development of the gut microbiota over time. After hospital discharge, infants exhibited a transition toward a microbiota community similar to that of adults by 3.5 years of age, in accordance with the functional metabolic pathways of the gut microbiota.IMPORTANCEThis study is among the very few reports analyzing the gut microbiota development in very preterm infants over time in a large, multicenter population of 596 children from a well-described nationwide birth cohort, with a follow-up until the age of 3.5 years. The maturation of the intestinal microbiota was confirmed to occur over time, with increased alpha diversity and decreased beta diversity. Specifically, 13 microbiota clusters were identified during the hospitalization period, while and only three clusters were observed at 3.5 years. Infants born prematurely or via Cesarean section exhibited a less stable microbiota, frequently shifting clusters. A number of perinatal factors were identified as influencing the development of the microbiota. Among these, the preconceptional maternal BMI emerged as the only consistent factor up to 3.5 years. The metabolic pathways of the microbiota evolved over time, in accordance with the maturation of the gut microbiota.
Collapse
Affiliation(s)
- Gaël Toubon
- INSERM, UMR1153 Centre de Recherche Épidémiologie et StatistiqueS (CRESS), Université Paris Cité, Paris, France
- INSERM, UMR-S 1139, Physiopathologie et Pharmacotoxicologie Placentaire Humaine Microbiote Pré & Postnatal, Université Paris Cité,, Paris, France
- FHU PREMA, “Fighting Prematurity”, Paris, France
| | - Constance Patin
- INRAE, UMR 1319, AgrosParisTech, Institut Micalis, Université Paris-Saclay, Paris, France
| | - Johanne Delannoy
- INSERM, UMR-S 1139, Physiopathologie et Pharmacotoxicologie Placentaire Humaine Microbiote Pré & Postnatal, Université Paris Cité,, Paris, France
- FHU PREMA, “Fighting Prematurity”, Paris, France
| | - Jean-Christophe Rozé
- INRAE, UMR 1280, Physiologie des Adaptations Nutritionnelles (PhAN), Université Hospitalière de Nantes, Nantes, France
| | - Frédéric Barbut
- INSERM, UMR-S 1139, Physiopathologie et Pharmacotoxicologie Placentaire Humaine Microbiote Pré & Postnatal, Université Paris Cité,, Paris, France
- FHU PREMA, “Fighting Prematurity”, Paris, France
| | - Pierre-Yves Ancel
- INSERM, UMR1153 Centre de Recherche Épidémiologie et StatistiqueS (CRESS), Université Paris Cité, Paris, France
- FHU PREMA, “Fighting Prematurity”, Paris, France
| | - Marie-Aline Charles
- INSERM, UMR1153 Centre de Recherche Épidémiologie et StatistiqueS (CRESS), Université Paris Cité, Paris, France
| | - Marie-José Butel
- INSERM, UMR-S 1139, Physiopathologie et Pharmacotoxicologie Placentaire Humaine Microbiote Pré & Postnatal, Université Paris Cité,, Paris, France
- FHU PREMA, “Fighting Prematurity”, Paris, France
| | - Patricia Lepage
- INRAE, UMR 1319, AgrosParisTech, Institut Micalis, Université Paris-Saclay, Paris, France
| | - Julio Aires
- INSERM, UMR-S 1139, Physiopathologie et Pharmacotoxicologie Placentaire Humaine Microbiote Pré & Postnatal, Université Paris Cité,, Paris, France
- FHU PREMA, “Fighting Prematurity”, Paris, France
| |
Collapse
|
8
|
Kim SH, Kwak M, Hwang JK, Keum J, Jin HY, Lee CY, Tanpure RS, Kim YJ, Hoh JK, Park JY, Chung W, Jeon BH, Park HK. Altered heme metabolism and hemoglobin concentration due to empirical antibiotics-induced gut dysbiosis in preterm infants. Comput Struct Biotechnol J 2025; 27:937-945. [PMID: 40123796 PMCID: PMC11930222 DOI: 10.1016/j.csbj.2025.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/04/2025] [Accepted: 03/04/2025] [Indexed: 03/25/2025] Open
Abstract
Background High-risk infants are usually treated with empirical antibiotics after birth, regardless of the evidence of infection; however, their gut microbiome and metabolome have seldom been studied. This study investigated the influence of antibiotic exposure on the gut microbiome and associated metabolic pathways in term and preterm infants. Methods Thirty-six infants within 10 days of birth who were admitted to a neonatal intensive care unit/newborn nursery unit were divided into four groups based on maturity (gestational age) and use of empirical antibiotics. Genomic DNA was extracted from the fecal samples and underwent high-throughput 16S rRNA amplicon sequencing using the Illumina platforms. Taxonomic classification, diversity analysis, and metagenomic function prediction were performed. Results Preterm infants with empirical antibiotics showed a significantly decreased population of Firmicutes (p = 0.003) and an increased population of Proteobacteria (p < 0.001) compared to other groups. At the genus level, the populations of Raoultella (p = 0.065) and Escherichia (p = 0.052) showed an increased trend. The change in microbial composition was correlated with increased heme biosynthesis and decreased hemoglobin levels. Conclusion Collectively, our finding suggested that empirical antibiotic exposure in preterm infants alters the gut microbiome, potentially leading to adverse health outcomes. This dysbiosis may affect heme metabolism, increasing the risk of anemia in these vulnerable infants. Therefore, antibiotic use should be carefully tailored to minimize potential harm.
Collapse
Affiliation(s)
- Seung Hyun Kim
- Department of Pediatrics, Hanyang University College of Medicine, Seoul 04763, South Korea
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea
| | - Min‑Jin Kwak
- Emerging Pathogens Institute, University of Florida, Gainesville, FL 32611, USA
| | - Jae Kyoon Hwang
- Department of Pediatrics, Hanyang University College of Medicine, Seoul 04763, South Korea
| | - Jihyun Keum
- Department of Obstetrics and Gynecology, Hanyang University College of Medicine, Seoul 04763, South Korea
| | - Hee Yeon Jin
- Division of Microbiome, Int-Gen Company, Seoul 04799, South Korea
| | - Chan-Yeong Lee
- Department of Earth Resources and Environmental Engineering, Hanyang University, Seoul 04763, South Korea
| | - Rahul Sadashiv Tanpure
- Department of Earth Resources and Environmental Engineering, Hanyang University, Seoul 04763, South Korea
| | - Yong Joo Kim
- Department of Pediatrics, Hanyang University College of Medicine, Seoul 04763, South Korea
| | - Jeong-Kyu Hoh
- Department of Obstetrics and Gynecology, Hanyang University College of Medicine, Seoul 04763, South Korea
| | - Jae Yong Park
- Division of Microbiome, Int-Gen Company, Seoul 04799, South Korea
| | - Woojin Chung
- Department of Civil & Energy System Engineering, Kyonggi University, Suwon 16227, South Korea
| | - Byong-Hun Jeon
- Department of Earth Resources and Environmental Engineering, Hanyang University, Seoul 04763, South Korea
| | - Hyun-Kyung Park
- Department of Pediatrics, Hanyang University College of Medicine, Seoul 04763, South Korea
| |
Collapse
|
9
|
McGann C, Phyu R, Bittinger K, Mukhopadhyay S. Role of the Microbiome in Neonatal Infection: Pathogenesis and Implications for Management. Clin Perinatol 2025; 52:147-166. [PMID: 39892949 DOI: 10.1016/j.clp.2024.10.010] [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] [Indexed: 02/04/2025]
Abstract
The human microbiome refers to the collective genome of microorganisms, including bacteria, fungi, and viruses residing on human body surfaces that are in contact with the environment. Together these communities protect against invasive infections. Conversely, when disrupted, the microbiome can be the source of pathogens causing invasive infection. Interventions to manipulate it via probiotics, antibiotics, and fecal transplantation are available. The risk benefit of these interventions remains unclear. In this review, the authors discuss evidence linking the gut microbiome to neonatal sepsis and also discuss the challenges for translating this knowledge into better clinical care.
Collapse
Affiliation(s)
- Carolyn McGann
- Division of Neonatology, Department of Pediatrics, Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA 19104, USA; Perelman School of Medicine, University of Pennsylvania, 3401 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Riley Phyu
- Department of Clinical Education and Assessment Center, Rown-Virtua School of Osteopathic Medicine, Rowan University, 1 Medical Center Drive Stratford, NJ 08084, USA
| | - Kyle Bittinger
- Perelman School of Medicine, University of Pennsylvania, 3401 Civic Center Boulevard, Philadelphia, PA 19104, USA; Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Sagori Mukhopadhyay
- Division of Neonatology, Department of Pediatrics, Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA 19104, USA; Perelman School of Medicine, University of Pennsylvania, 3401 Civic Center Boulevard, Philadelphia, PA 19104, USA.
| |
Collapse
|
10
|
Fu Z, Jiang Z, Zhao F, Gou T, Jiang L. Causal association between gastroesophageal reflux disease and sepsis, and the mediating role of gut bacterial abundance, a Mendelian randomization study. Medicine (Baltimore) 2025; 104:e41631. [PMID: 39993106 PMCID: PMC11857025 DOI: 10.1097/md.0000000000041631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 02/26/2025] Open
Abstract
Gastroesophageal reflux disease (GERD), akin to sepsis, is mediated by inflammatory reactions and exhibits a strong correlation with intestinal dysbiosis. We sought to examine whether these associations reflect causality using the Mendelian randomization (MR) mediation analysis. Genetic data were obtained from genome-wide association studies. Two-sample MR were performed to evaluate the causal association, accompanied by sensitivity analyses. Reverse direction MR was undertaken to assess the potential for reverse causation. Then, mediation analysis was performed to evaluate the mediating effect of gut bacterial pathway abundance in this relationship. Genetic predisposition to GERD was significantly associated with sepsis [inverse variance weighting: odds ratio = 1.366, P = 2.13E-09, 95% confidence interval [CI] 1.233-1.513] and sepsis-related 28-day mortality (inverse variance weighting: odds ratio = 1.412, P = 6.64E-03, 95% CI 1.101-1.812). There is no convincing evidence for reverse causation. Gut bacterial pathway abundance (ARO.PWY..chorismate.biosynthesis.I) mediates the effect of GERD on sepsis (β = 0.036, 95% CI 0.004-0.067, P = .025), accounting for 11.406% of the total effect; Gut bacterial pathway abundance (PWY.7219..adenosine.ribonucleotides.de.novo.biosynthesis) mediates the effect of GERD on sepsis (β = 0.026, 95% CI -0.003 to 0.056, P = .083), accounting for 8.486% of the total effect; gut bacterial pathway abundance (ARO.PWY..chorismate.biosynthesis.I) mediates the effect of GERD on sepsis (28-day death) (β = 0.079, 95% CI 0.005-0.153, P = .036), accounting for 22.890% of the total effect; gut bacterial pathway abundance (TRNA.CHARGING.PWY..tRNA.charging) mediates the effect of GERD on sepsis (28-day death) (β = -0.066, 95% CI -0.140 to 0.007, P = .078), accounting for -19.171% of the total effect. The present MR study supported GERD as a causal risk factor of sepsis and sepsis-related 28-day mortality. Three specific gut bacterial pathway abundances were identified that played a partial mediating role in the aforementioned causal relationship between GERD and sepsis.
Collapse
Affiliation(s)
- Zengyan Fu
- Department of Emergency Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Zhenhong Jiang
- Department of Emergency Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Fei Zhao
- Department of Emergency Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Tao Gou
- Department of Emergency Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Le Jiang
- Department of Emergency Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
11
|
Wang H, Chen D, Li H, Fu C, Fang L, Wang R, Xu J. Bifidobacterium regulates premature infant gut metabolites, reducing serum inflammatory factors: a randomised controlled trial. Pediatr Res 2025; 97:1171-1182. [PMID: 39271904 DOI: 10.1038/s41390-024-03552-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 08/17/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND Analyse the effects of Bifidobacterium BB-12 on intestinal metabolites and serum inflammatory factors in premature infants. METHODS 71 premature infants at gestational age of ≤32 weeks were randomly divided into the probiotic (n = 36) and control (n = 35) groups. Faecal and blood samples were collected from the two groups of premature infants at the 2nd and 4th week of life for intestinal metabolite detection and assessment of the level of the serum inflammatory markers TLR4, NF- κ B, IL-1β, and TNF- α. RESULTS Compared to the control group, the probiotic group contained more amino acids, these elements were enriched on multiple amino acid metabolic pathways, and the probiotic group showed significantly lower levels of the serum inflammatory markers TLR4, NF-κB, IL-1β, and TNF-α. Finally, the probiotic group showed a lower incidence of feeding intolerance. CONCLUSIONS The administration of Bifidobacterium BB-12 is associated with increasing the levels of glutamine, glutamic acid, and kynurenine in the gut of premature infants, and associated with reducing the levels of TLR4 and NF-κB in the serum, further decreasing the secretion of the pro-inflammatory factors IL-1β and TNF-α, and alleviating systemic inflammatory reactions, thereby reducing the incidence of feeding intolerance. IMPACT 1. The use of Bifidobacterium BB-12 in premature infants can increase the levels of amino acids in the intestine. 2. Increases in Bifidobacterium BB-12 may decrease the serum levels of TLR4, NF-κB, IL-1β, and TNF-α. 3. Kynurenine may improve the prognosis of preterm infants by reducing inflammation. 4. Bifidobacterium BB-12 may improve the feeding tolerance of premature infants, thus reducing the incidence of feeding intolerance.
Collapse
Affiliation(s)
- He Wang
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China
- Department of Neonatology, Quanzhou Maternity and Children's Hospital, Quanzhou, Fujian Province, China
| | - Dongmei Chen
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China
- Department of Neonatology, Quanzhou Maternity and Children's Hospital, Quanzhou, Fujian Province, China
| | - Huamei Li
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China
- Department of Neonatology, Quanzhou Maternity and Children's Hospital, Quanzhou, Fujian Province, China
| | - Chunyan Fu
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China
- Department of Neonatology, Quanzhou Maternity and Children's Hospital, Quanzhou, Fujian Province, China
| | - Lingyu Fang
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China
- Department of Neonatology, Quanzhou Maternity and Children's Hospital, Quanzhou, Fujian Province, China
| | - Ruiquan Wang
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China
- Department of Neonatology, Quanzhou Maternity and Children's Hospital, Quanzhou, Fujian Province, China
| | - Jinglin Xu
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China.
- Department of Neonatology, Quanzhou Maternity and Children's Hospital, Quanzhou, Fujian Province, China.
| |
Collapse
|
12
|
Renk H, Schoppmeier U, Müller J, Kuger V, Neunhoeffer F, Gille C, Peter S. Oxygenation and intestinal perfusion and its association with perturbations of the early life gut microbiota composition of children with congenital heart disease. Front Microbiol 2025; 15:1468842. [PMID: 39881980 PMCID: PMC11775010 DOI: 10.3389/fmicb.2024.1468842] [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: 07/22/2024] [Accepted: 12/10/2024] [Indexed: 01/31/2025] Open
Abstract
Background Early life gut microbiota is known to shape the immune system and has a crucial role in immune homeostasis. Only little is known about composition and dynamics of the intestinal microbiota in infants with congenital heart disease (CHD) and potential influencing factors. Methods We evaluated the intestinal microbial composition of neonates with CHD (n = 13) compared to healthy controls (HC, n = 30). Fecal samples were analyzed by shotgun metagenomics. Different approaches of statistical modeling were applied to assess the impact of influencing factors on variation in species composition. Unsupervised hierarchical clustering of the microbial composition of neonates with CHD was used to detect associations of distinct clusters with intestinal tissue oxygenation and perfusion parameters, obtained by the "oxygen to see" (O2C) method. Results Overall, neonates with CHD showed an intestinal core microbiota dominated by the genera Enterococcus (27%) and Staphylococcus (20%). Furthermore, a lower abundance of the genera Bacteroides (8% vs. 14%), Parabacteroides (1% vs. 3%), Bifidobacterium (4% vs. 12%), and Escherichia (8% vs. 23%) was observed in CHD compared to HCs. CHD patients that were born by vaginal delivery showed a lower fraction of the genera Bacteroides (15% vs. 21%) and Bifidobacterium (7% vs. 22%) compared to HCs and in those born by cesarean section, these genera were not found at all. In infants with CHD, we found a significant impact of oxygen saturation (SpO2) on relative abundances of the intestinal core microbiota by multivariate analysis of variance (F[8,2] = 24.9, p = 0.04). Statistical modeling suggested a large proportional shift from a microbiota dominated by the genus Streptococcus (50%) in conditions with low SpO2 towards the genus Enterococcus (61%) in conditions with high SpO2. We identified three distinct compositional microbial clusters, corresponding neonates differed significantly in intestinal blood flow and global gut perfusion. Conclusion Early life differences in gut microbiota of CHD neonates versus HCs are possibly linked to oxygen levels. Delivery method may affect microbiota stability. However, further studies are needed to assess the effect of potential interventions including probiotics or fecal transplants on early life microbiota perturbations in neonates with CHD.
Collapse
Affiliation(s)
- Hanna Renk
- Department of Neuropediatrics, Developmental Neurology and Social Pediatrics, University Children’s Hospital Tübingen, Tübingen, Germany
- German Centre for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
- Department of Pediatric Cardiology, Pulmonology and Pediatric Intensive Care Medicine, University Children’s Hospital Tübingen, Tübingen, Germany
| | - Ulrich Schoppmeier
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Tübingen, Germany
| | - Jennifer Müller
- NGS Competence Center Tübingen (NCCT), University of Tübingen, Tübingen, Germany
| | - Vanessa Kuger
- Department of Pediatric Cardiology, Pulmonology and Pediatric Intensive Care Medicine, University Children’s Hospital Tübingen, Tübingen, Germany
| | - Felix Neunhoeffer
- Department of Pediatric Cardiology, Pulmonology and Pediatric Intensive Care Medicine, University Children’s Hospital Tübingen, Tübingen, Germany
| | - Christian Gille
- Department of Neonatology, Heidelberg University Children’s Hospital, Heidelberg, Germany
| | - Silke Peter
- German Centre for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Tübingen, Germany
| |
Collapse
|
13
|
Zeng S, Zhou M, Mu D, Wang S. Clinical implications of maternal multikingdom transmissions and early-life microbiota. THE LANCET. MICROBE 2025:101042. [PMID: 39818230 DOI: 10.1016/j.lanmic.2024.101042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 11/04/2024] [Accepted: 11/12/2024] [Indexed: 01/18/2025]
Abstract
Mother-to-infant transmission of the bacteriome, virome, mycobiome, archaeome, and their mobile genetic elements has been recognised in nature as an important step for the infant to acquire and maintain a healthy early-life (from birth till age 3 years) microbiota. A comprehensive overview of other maternal multikingdom transmissions remains unavailable, except for that of the bacteriome. Associations between microorganisms and diseases throughout the human life span have been gradually discovered; however, whether these microorganisms are maternally derived and how they concomitantly interact with other microbial counterparts remain poorly understood. This Review first discusses the current understanding of maternal multikingdom transmissions, their contributions to the development of early-life microbiota, and the primary factors that influence the transmission processes. The clinical implications of the inherited microbiota on human health in early life have been emphasised upon next, along with highlighting of knowledge gaps that should be addressed in future research. Finally, interventions to restore typical vertical transmission or disturbed early-life microbiota have been discussed as potential therapeutic approaches.
Collapse
Affiliation(s)
- Shuqin Zeng
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Meicen Zhou
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Dezhi Mu
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China.
| | - Shaopu Wang
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
14
|
Almási ÉDH, Eisenhard L, Osbelt L, Lesker TR, Vetter AC, Knischewski N, Bielecka AA, Gronow A, Muthukumarasamy U, Wende M, Tawk C, Neumann-Schaal M, Brönstrup M, Strowig T. Klebsiella oxytoca facilitates microbiome recovery via antibiotic degradation and restores colonization resistance in a diet-dependent manner. Nat Commun 2025; 16:551. [PMID: 39789003 PMCID: PMC11717976 DOI: 10.1038/s41467-024-55800-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025] Open
Abstract
Competition among bacteria for carbohydrates is pivotal for colonization resistance (CR). However, the impact of Western-style diets on CR remains unclear. Here we show how the competition between Klebsiella oxytoca and Klebsiella pneumoniae is modulated by consuming one of three Western-style diets characterized by high-starch, high-sucrose, or high-fat/high-sucrose content. In vivo competition experiments in ampicillin-treated mice reveal that K. oxytoca promotes K. pneumoniae decolonization on all dietary backgrounds. However, mice on the high-fat/high-sucrose diet show reduced pathogen clearance. Microbiome analysis reveals that the combination of Western-style diets and ampicillin treatment synergize in microbiome impairment, particularly noticeable in the presence of high dietary fat content. The diet-independent degradation of ampicillin in the gut lumen by K. oxytoca beta-lactamases facilitates rapid commensal outgrowth, which is required for subsequent pathogen clearance. Our findings provide insights into how diet modulates functional microbiome recovery and K. oxytoca-mediated pathogen elimination from the gut.
Collapse
Affiliation(s)
- Éva D H Almási
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Lea Eisenhard
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Lisa Osbelt
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Till Robin Lesker
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Anna C Vetter
- Department of Chemical Biology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Nele Knischewski
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Agata Anna Bielecka
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Achim Gronow
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Uthayakumar Muthukumarasamy
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Marie Wende
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Caroline Tawk
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Meina Neumann-Schaal
- Bacterial Metabolomics, Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Mark Brönstrup
- Department of Chemical Biology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Till Strowig
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany.
- Center for Individualized Infection Medicine, Hannover, Germany.
- German Center for Infection Research (DZIF), partner site Hannover-Braunschweig, Braunschweig, Germany.
| |
Collapse
|
15
|
Vaher K, Cabez MB, Parga PL, Binkowska J, van Beveren GJ, Odendaal ML, Sullivan G, Stoye DQ, Corrigan A, Quigley AJ, Thrippleton MJ, Bastin ME, Bogaert D, Boardman JP. The neonatal gut microbiota: A role in the encephalopathy of prematurity. Cell Rep Med 2024; 5:101845. [PMID: 39637857 PMCID: PMC11722115 DOI: 10.1016/j.xcrm.2024.101845] [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: 09/27/2023] [Revised: 07/11/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024]
Abstract
Preterm birth correlates with brain dysmaturation and neurocognitive impairment. The gut microbiome associates with behavioral outcomes in typical development, but its relationship with neurodevelopment in preterm infants is unknown. We characterize fecal microbiome in a cohort of 147 neonates enriched for very preterm birth using 16S-based and shotgun metagenomic sequencing. Delivery mode strongly correlates with the preterm microbiome shortly after birth. Low birth gestational age, infant sex assigned at birth, and antibiotics associate with microbiome composition at neonatal intensive care unit discharge. We integrate these data with term-equivalent structural and diffusion brain MRI. Bacterial community composition associates with MRI features of encephalopathy of prematurity. Particularly, abundances of Escherichia coli and Klebsiella spp. correlate with microstructural parameters in deep and cortical gray matter. Metagenome functional capacity analyses suggest that these bacteria may interact with brain microstructure via tryptophan and propionate metabolism. This study indicates that the gut microbiome associates with brain development following preterm birth.
Collapse
Affiliation(s)
- Kadi Vaher
- Centre for Reproductive Health, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK; Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Manuel Blesa Cabez
- Centre for Reproductive Health, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK; Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Paula Lusarreta Parga
- Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Justyna Binkowska
- Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Gina J van Beveren
- Department of Pediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital and University Medical Center Utrecht, 3584 EA Utrecht, the Netherlands
| | - Mari-Lee Odendaal
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven 3721 MA, the Netherlands
| | - Gemma Sullivan
- Centre for Reproductive Health, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK; Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - David Q Stoye
- Centre for Reproductive Health, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Amy Corrigan
- Centre for Reproductive Health, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Alan J Quigley
- Department of Paediatric Radiology, Royal Hospital for Children and Young People, Edinburgh EH16 4TJ, UK
| | | | - Mark E Bastin
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Debby Bogaert
- Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK; Department of Pediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital and University Medical Center Utrecht, 3584 EA Utrecht, the Netherlands
| | - James P Boardman
- Centre for Reproductive Health, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK; Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
16
|
Cantón R, De Lucas Ramos P, García-Botella A, García-Lledó A, Hernández-Sampelayo T, Gómez-Pavón J, González Del Castillo J, Martín-Delgado MC, Martín Sánchez FJ, Martínez-Sellés M, Molero García JM, Moreno Guillén S, Rodríguez-Artalejo FJ, Reigadas E, Del Campo R, Serrano S, Ruiz-Galiana J, Bouza E. Human intestinal microbiome: Role in health and disease. REVISTA ESPANOLA DE QUIMIOTERAPIA : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE QUIMIOTERAPIA 2024; 37:438-453. [PMID: 38978509 PMCID: PMC11578434 DOI: 10.37201/req/056.2024] [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: 06/21/2024] [Accepted: 07/01/2024] [Indexed: 07/10/2024]
Abstract
The study of the microbiota and the microbiome, and specifically the intestinal one, has determined great interest due to the possible association of their alterations with numerous diseases. These include entities as diverse as Crohn's disease, autism, diabetes, cancer or situations as prevalent today as obesity. In view of this situation, different recommendations have been performed regarding the use of probiotics, prebiotics, and postbiotics as modulators of the microbiota and the microbiome, seeking both preventive and therapeutic effects, and faecal material transfer (FMT) is proposed as an alternative. The latter has emerged as the only proven beneficial intervention on the intestinal microbiome, specifically in the treatment of recurrent colitis associated with Clostridioides difficile (R-CDI). In the rest of the entities, the lowering of laboratory costs has favored the study of the microbiome, which is resolved by delivering reports with catalogs of microorganisms, metabolites or supposed biomarkers without consensus on their composition associated with healthy or diseased microbiota and the disease. There is still insufficient evidence in any disease for interventions on the microbiome beyond FMT and R-CDI. Multi- and multi-disciplinary work with extensive research and the application of artificial intelligence in this field may shed light on the questions raised currently. Ethical issues must also be resolved in light of possible interventions within the umbrella of personalized medicine.
Collapse
Affiliation(s)
- R Cantón
- Rafael Cantón. Servicio de Microbiología. Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria. CIBER de Enfermedades Infecciosas (CIBERINFEC). Madrid, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Graspeuntner S, Lupatsii M, van Zandbergen V, Dammann MT, Pagel J, Nguyen DN, Humberg A, Göpel W, Herting E, Rupp J, Härtel C, Fortmann I. Infants < 90 days of age with late-onset sepsis display disturbances of the microbiome-immunity interplay. Infection 2024:10.1007/s15010-024-02396-6. [PMID: 39541036 DOI: 10.1007/s15010-024-02396-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 09/09/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVE We hypothesized that previously healthy infants < 90 days of age with late-onset sepsis (LOS) have disturbances of the gut microbiome with yet undefined specific immunological patterns. METHODS We performed a prospective single-center convenience sample study between January 2019 and July 2021 in a case-control design. Routine diagnostics included conventional cultures (blood, cerebrospinal fluid, urine), PCRs and inflammatory markers in infants aged < 90 days with clinical LOS. We additionally analyzed blood lymphocyte subsets including CD4 + CD25 + forkhead box protein (FoxP3)+ Tregs and performed 16 S rRNA sequencing of stool samples, both compared to age-matched healthy controls. Results were adjusted for potential confounders that may influence microbial composition. RESULTS 51 infants with fever and clinical LOS were enrolled. Bacterial sepsis was diagnosed in n = 24 (47.1%) and viral infection in n = 13 (25.5%) infants, whereas in 14 (27.3%) infants the cause of fever remained undetermined. When compared to healthy controls, the gut microbiome of LOS infants at disease onset was characterized by a shift in community composition, specifically, decreased abundance of B. longum and an increase of Bacteroidia spp. Intriguingly, the abundance of B. longum negatively correlated with the frequency of blood CD4-positive cells in healthy controls but not in infants with LOS. At one year of age, we observed microbiome differences in infants with history of LOS when compared to healthy controls, such as an increased gut microbial diversity. CONCLUSION Our data suggest potential signatures of the microbiome-immunity interplay in infants with LOS, which should be investigated further as possible targets for prevention.
Collapse
Affiliation(s)
- Simon Graspeuntner
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Lübeck, Germany
| | - Mariia Lupatsii
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Vera van Zandbergen
- Department of Pediatrics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Marie-Theres Dammann
- Department of Pediatrics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Julia Pagel
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Lübeck, Germany
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Duc Ninh Nguyen
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Wolfgang Göpel
- Department of Pediatrics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Egbert Herting
- Department of Pediatrics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Jan Rupp
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Lübeck, Germany
| | - Christoph Härtel
- Department of Pediatrics, University Hospital of Würzburg, Würzburg, Germany
| | - Ingmar Fortmann
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Lübeck, Germany.
- Department of Pediatrics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany.
| |
Collapse
|
18
|
Xuan H, Umar S, Zhong C, Yu W, Ahmed I, Wheatley JL, Sampath V, Chavez-Bueno S. Lactobacillus rhamnosus modulates murine neonatal gut microbiota and inflammation caused by pathogenic Escherichia coli. BMC Microbiol 2024; 24:452. [PMID: 39506682 PMCID: PMC11539828 DOI: 10.1186/s12866-024-03598-6] [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: 06/19/2024] [Accepted: 10/23/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Pathogenic Escherichia coli strains produce neonatal septicemia after colonizing the neonatal gut. While the probiotic Lactobacillus rhamnosus GG (LGG) effectively reduces neonatal sepsis, LGG's effects on the neonatal intestinal microbiota alterations and inflammation triggered by E. coli are incompletely understood. We hypothesized that LGG significantly modulates the specific neonatal gut microbial populations changes and the inflammatory response elicited by the enteral introduction of septicemia-producing E. coli. To test this hypothesis, newborn rats were pretreated orally with LGG or placebo prior to infection with the neonatal E. coli septicemia clinical isolate SCB34. Amplicon 16S rRNA gene sequencing was performed on intestinal samples. Intestinal injury and expression of inflammatory mediators and apoptosis were determined. RESULTS Alpha diversity of gut microbiota was greater in SCB34-infected pups in comparison to sham-infected pups, these changes were not modified by LGG pretreatment. Beta diversity analyses also showed differences between SCB34-infected vs. uninfected pups. LGG pretreatment before SCB34 infection did not result in significant beta diversity changes compared to placebo. Moreover, individual genera and species abundance analyses by linear discriminant analysis effect size (LEfSe) showed significant changes in Gram-negative, Gram-positive, and anaerobic populations resulting from LGG pretreatment and SCB34 infection. LGG significantly suppressed the expression of inflammatory cytokines but did not attenuate SCB34-induced apoptosis or histologic injury. CONCLUSIONS LGG modulates clinically significant microbiota features and inflammation triggered by pathogenic E. coli intestinal infection shortly after birth. This new knowledge can potentially be harnessed to design novel interventions against gut-derived neonatal sepsis.
Collapse
Affiliation(s)
- Hao Xuan
- Department of Electrical Engineering and Computer Science, University of Kansas, Lawrence, KS, USA
| | - Shahid Umar
- Department of Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| | - Cuncong Zhong
- Department of Electrical Engineering and Computer Science, University of Kansas, Lawrence, KS, USA
| | - Wei Yu
- Department of Pediatrics, Division of Neonatology, Children's Mercy Kansas City, Kansas City, MO, USA
| | - Ishfaq Ahmed
- Department of Math, Science and Computer Technology, Kansas City Kansas Community College, Kansas City, KS, USA
| | - Joshua L Wheatley
- Department of Pediatrics, Division of Infectious Diseases, Children's Mercy Kansas City, Kansas City, MO, USA
| | - Venkatesh Sampath
- Department of Pediatrics, Division of Neonatology, Children's Mercy Kansas City, Kansas City, MO, USA
- University of Missouri Kansas City School of Medicine, 2401 Gillham Road, Kansas City, MO, 64108, USA
| | - Susana Chavez-Bueno
- Department of Pediatrics, Division of Infectious Diseases, Children's Mercy Kansas City, Kansas City, MO, USA.
- University of Missouri Kansas City School of Medicine, 2401 Gillham Road, Kansas City, MO, 64108, USA.
| |
Collapse
|
19
|
Liu L, Ma L, Liu H, Zhao F, Li P, Zhang J, Lü X, Zhao X, Yi Y. Targeted discovery of gut microbiome-remodeling compounds for the treatment of systemic inflammatory response syndrome. mSystems 2024; 9:e0078824. [PMID: 39235366 PMCID: PMC11494991 DOI: 10.1128/msystems.00788-24] [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: 06/12/2024] [Accepted: 07/09/2024] [Indexed: 09/06/2024] Open
Abstract
Systemic inflammatory response syndrome (SIRS) is a severe inflammatory response that can lead to organ dysfunction and death. Modulating the gut microbiome is a promising therapeutic approach for managing SIRS. This study assesses the therapeutic potential of the Xuanfei Baidu (XFBD) formula in treating SIRS. The results showed that XFBD administration effectively reduced mortality rates and inflammation in SIRS mice. Using 16S rRNA sequencing and fecal microbiota transplantation (FMT), we substantiated that the therapeutic effects of XFBD are partly attributed to gut microbiota modulation. We conducted in vitro experiments to accurately assess the gut microbiome remodeling effects of 51 compounds isolated from XFBD. These compounds exhibited varying abilities to induce a microbial structure that closely resembles that of the healthy control group. By quantifying their impact on microbial structure and clustering their regulatory patterns, we devised multiple gut microbiome remodeling compound (GMRC) cocktails. GMRC cocktail C, comprising aucubin, gentiopicroside, syringic acid, gallic acid, p-hydroxybenzaldehyde, para-hydroxybenzoic acid, and isoimperatorin, demonstrated superior efficacy in treating SIRS compared to a single compound or to other cocktails. Finally, in vitro experiments showcased that GMRC cocktail C effectively rebalanced bacteria composition in SIRS patients. This study underscores XFBD's therapeutic potential in SIRS and highlights the importance of innovative treatment approaches for this disease by targeting the gut microbiota.IMPORTANCEDeveloping effective treatment strategies for systemic inflammatory response syndrome (SIRS) is crucial due to its severe and often life-threatening nature. While traditional treatments like dexamethasone have shown efficacy, they also come with significant side effects and limitations. This study makes significant strides by demonstrating that the Xuanfei Baidu (XFBD) formula can substantially reduce mortality rates and inflammation in SIRS mice through effective modulation of the gut microbiota. By quantitatively assessing the impact of 51 compounds derived from XFBD on the gut microbiome, we developed a potent gut microbiome remodeling compound cocktail. This cocktail outperformed individual compounds and other mixtures in efficacy against SIRS. These findings highlight the potential of XFBD as a therapeutic solution for SIRS and underscore the critical role of innovative strategies targeting the gut microbiota in addressing this severe inflammatory condition.
Collapse
Affiliation(s)
- Luyao Liu
- College of Food Science and Engineering, Northwest A&F University, Shaanxi, China
| | - Lin Ma
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Huan Liu
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fan Zhao
- College of Animal Science and Technology, Northwest A&F University, Shaanxi, China
| | - Pu Li
- Department of Critical Care Medicine, The Second Affiliated Hospital of Air Force Medical University, China, Shaanxi
| | - Junhua Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xin Lü
- College of Food Science and Engineering, Northwest A&F University, Shaanxi, China
| | - Xin Zhao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yanglei Yi
- College of Food Science and Engineering, Northwest A&F University, Shaanxi, China
| |
Collapse
|
20
|
Chae H, Kim SY, Kang HM, Im SA, Youn YA. Dysbiosis of the initial stool microbiota increases the risk of developing necrotizing enterocolitis or feeding intolerance in newborns. Sci Rep 2024; 14:24416. [PMID: 39424878 PMCID: PMC11489565 DOI: 10.1038/s41598-024-75157-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 10/03/2024] [Indexed: 10/21/2024] Open
Abstract
Several perinatal factors influence the intestinal microbiome of newborns during the first days of life, whether during delivery or even in utero. These factors may increase the risk of developing necrotizing enterocolitis (NEC) by causing dysbiosis linked to a NEC-associated microbiota, which may also be associated with other gastrointestinal problems. The objective of our study was to evaluate the potential risks associated with microbial shifts in newborns with gastrointestinal symptoms and identify the intestinal microbiota of neonates at risk for NEC.During the study period, 310 preterm and term newborns' first passed meconium occurring within 72 h of birth were collected, and the microbiome was analyzed. We identified the risk factors in the NEC/FI group. Regarding microbiota, we compared the bacterial abundance between the NEC/FI group at the phylum and genus levels and explored the differences in the microbial composition of the 1st stool samples. A total of 14.8% (n = 46) of the infants were diagnosed with NEC or FI. In univariate analysis, the mean gestational age and birth weight were significantly lower in the NEC/FI group (p < 0.001). Prolonged rupture of membranes (PROM) > 18 h, chorioamnionitis, and histology were significantly higher in the NEC/FI group (p < 0.001). Multivariate analysis showed that gestational age (GA), prolonged membrane rupture (> 18 h), and early onset sepsis were consistently associated with an increased risk of NEC/FI. Infants diagnosed with NEC/FI exhibited a significantly lower abundance of Actinobacteria at the phylum level than the control group (p < 0.001). At the genus level, a significantly lower abundance of Streptococcus and Bifidobacterium which belong to the Actinobacteria phylum, was observed in the NEC/FI group (p < 0.001). Furthermore, the NEC/FI had significantly lower alpha diversities (Shannon Index,3.39 vs. 3.12; P = 0.044, respectively). Our study revealed that newborns with lower diversity and dysbiosis in their initial gut microbiota had an increased risk of developing NEC, with microbiota differences appearing to be associated with NEC/FI. Dysbiosis could potentially serve as a predictive marker for NEC- or GI-related symptoms.
Collapse
Affiliation(s)
- Hyojin Chae
- Department of Laboratory Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Sae Yun Kim
- Department of Pediatrics, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Hyun Mi Kang
- Department of Pediatrics, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Soo-Ah Im
- Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Young-Ah Youn
- Department of Pediatrics, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
21
|
Thänert R, Schwartz DJ, Keen EC, Hall-Moore C, Wang B, Shaikh N, Ning J, Rouggly-Nickless LC, Thänert A, Ferreiro A, Fishbein SRS, Sullivan JE, Radmacher P, Escobedo M, Warner BB, Tarr PI, Dantas G. Clinical sequelae of gut microbiome development and disruption in hospitalized preterm infants. Cell Host Microbe 2024; 32:1822-1837.e5. [PMID: 39197454 PMCID: PMC11466706 DOI: 10.1016/j.chom.2024.07.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/24/2024] [Accepted: 07/31/2024] [Indexed: 09/01/2024]
Abstract
Aberrant preterm infant gut microbiota assembly predisposes to early-life disorders and persistent health problems. Here, we characterize gut microbiome dynamics over the first 3 months of life in 236 preterm infants hospitalized in three neonatal intensive care units using shotgun metagenomics of 2,512 stools and metatranscriptomics of 1,381 stools. Strain tracking, taxonomic and functional profiling, and comprehensive clinical metadata identify Enterobacteriaceae, enterococci, and staphylococci as primarily exploiting available niches to populate the gut microbiome. Clostridioides difficile lineages persist between individuals in single centers, and Staphylococcus epidermidis lineages persist within and, unexpectedly, between centers. Collectively, antibiotic and non-antibiotic medications influence gut microbiome composition to greater extents than maternal or baseline variables. Finally, we identify a persistent low-diversity gut microbiome in neonates who develop necrotizing enterocolitis after day of life 40. Overall, we comprehensively describe gut microbiome dynamics in response to medical interventions in preterm, hospitalized neonates.
Collapse
Affiliation(s)
- Robert Thänert
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Drew J Schwartz
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center for Women's Infectious Diseases Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Eric C Keen
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carla Hall-Moore
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Bin Wang
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nurmohammad Shaikh
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jie Ning
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - Anna Thänert
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Aura Ferreiro
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Skye R S Fishbein
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Janice E Sullivan
- Department of Pediatrics, University of Louisville School of Medicine, Norton Children's Hospital, Louisville, KY 40202, USA
| | - Paula Radmacher
- Department of Pediatrics, University of Louisville School of Medicine, Norton Children's Hospital, Louisville, KY 40202, USA
| | - Marilyn Escobedo
- Department of Pediatrics, University of Oklahoma, Oklahoma City, OK 73104, USA
| | - Barbara B Warner
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Phillip I Tarr
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Gautam Dantas
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA.
| |
Collapse
|
22
|
Ganjali M, Baghfalaki T, Balakrishnan N. Joint modeling of zero-inflated longitudinal measurements and time-to-event outcomes with applications to dynamic prediction. Stat Methods Med Res 2024; 33:1731-1767. [PMID: 39373068 DOI: 10.1177/09622802241268466] [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] [Indexed: 10/08/2024]
Abstract
In this article, we present a joint modeling approach for zero-inflated longitudinal count measurements and time-to-event outcomes. For the longitudinal sub-model, a mixed effects Hurdle model is utilized, incorporating various distributional assumptions such as zero-inflated Poisson, zero-inflated negative binomial, or zero-inflated generalized Poisson. For the time-to-event sub-model, a Cox proportional hazard model is applied. For the functional form linking the longitudinal outcome history to the hazard of the event, a linear combination is used. This combination is derived from the current values of the linear predictors of Hurdle mixed effects. Some other forms are also considered, including a linear combination of the current slopes of the linear predictors of Hurdle mixed effects as well as the shared random effects. A Markov chain Monte Carlo method is implemented for Bayesian parameter estimation. Dynamic prediction using joint modeling is highly valuable in personalized medicine, as discussed here for joint modeling of zero-inflated longitudinal count measurements and time-to-event outcomes. We assess and demonstrate the effectiveness of the proposed joint models through extensive simulation studies, with a specific emphasis on parameter estimation and dynamic predictions for both over-dispersed and under-dispersed data. We finally apply the joint model to longitudinal microbiome pregnancy and HIV data sets.
Collapse
Affiliation(s)
- Mojtaba Ganjali
- Department of Statistics, Faculty of Mathematical Sciences, Shahid Beheshti University, Tehran, Iran
| | - Taban Baghfalaki
- Inserm, Bordeaux Population Health Research Center (BPH) U1219, University of Bordeaux, ISPED, Bordeaux, France
| | | |
Collapse
|
23
|
Shama S, Asbury MR, Kiss A, Bando N, Butcher J, Comelli EM, Copeland JK, Greco A, Kothari A, Sherman PM, Stintzi A, Taibi A, Tomlinson C, Unger S, Wang PW, O'Connor DL. Mother's milk microbiota is associated with the developing gut microbial consortia in very-low-birth-weight infants. Cell Rep Med 2024; 5:101729. [PMID: 39243753 PMCID: PMC11525026 DOI: 10.1016/j.xcrm.2024.101729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/30/2024] [Accepted: 08/16/2024] [Indexed: 09/09/2024]
Abstract
Mother's milk contains diverse bacterial communities, although their impact on microbial colonization in very-low-birth-weight (VLBW, <1,500 g) infants remains unknown. Here, we examine relationships between the microbiota in preterm mother's milk and the VLBW infant gut across initial hospitalization (n = 94 mother-infant dyads, 422 milk-stool pairs). Shared zero-radius operational taxonomic units (zOTUs) between milk-stool pairs account for ∼30%-40% of zOTUs in the VLBW infant's gut. We show dose-response relationships between intakes of several genera from milk and their concentrations in the infant's gut. These relationships and those related to microbial sharing change temporally and are modified by in-hospital feeding practices (especially direct breastfeeding) and maternal-infant antibiotic use. Correlations also exist between milk and stool microbial consortia, suggesting that multiple milk microbes may influence overall gut communities together. These results highlight that the mother's milk microbiota may shape the gut colonization of VLBW infants by delivering specific bacteria and through intricate microbial interactions.
Collapse
Affiliation(s)
- Sara Shama
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada; Translational Medicine Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
| | - Michelle R Asbury
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada; Departments of Physiology & Pharmacology, and Pediatrics, University of Calgary, Calgary, AB, Canada
| | - Alex Kiss
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada; Evaluative and Clinical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Nicole Bando
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada; Translational Medicine Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
| | - James Butcher
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada; Ottawa Institute of Systems Biology, Ottawa, ON, Canada
| | - Elena M Comelli
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada; Joannah and Brian Lawson Centre for Child Nutrition, University of Toronto, Toronto, ON, Canada
| | - Julia K Copeland
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, ON, Canada
| | - Adrianna Greco
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada; Translational Medicine Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
| | - Akash Kothari
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada; Translational Medicine Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
| | - Philip M Sherman
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada; Cell Biology Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, ON, Canada; Gastroenterology, Hepatology, and Nutrition, The Hospital for Sick Children, Toronto, ON, Canada; Department of Paediatrics, University of Toronto, Toronto, ON, Canada
| | - Alain Stintzi
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada; Ottawa Institute of Systems Biology, Ottawa, ON, Canada
| | - Amel Taibi
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - Christopher Tomlinson
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada; Translational Medicine Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, ON, Canada; Joannah and Brian Lawson Centre for Child Nutrition, University of Toronto, Toronto, ON, Canada; Department of Paediatrics, University of Toronto, Toronto, ON, Canada; Division of Neonatology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Sharon Unger
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada; Department of Paediatrics, University of Toronto, Toronto, ON, Canada; Division of Neonatology, Izaak Walton Killam Hospital, Halifax, NS, Canada; Department of Pediatrics, Sinai Health, Toronto, ON, Canada; Rogers Hixon Ontario Human Milk Bank, Toronto, ON, Canada
| | - Pauline W Wang
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, ON, Canada
| | - Deborah L O'Connor
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada; Translational Medicine Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, ON, Canada; Joannah and Brian Lawson Centre for Child Nutrition, University of Toronto, Toronto, ON, Canada; Department of Pediatrics, Sinai Health, Toronto, ON, Canada; Rogers Hixon Ontario Human Milk Bank, Toronto, ON, Canada.
| |
Collapse
|
24
|
Lai MY, Chang YH, Lee CC. The impact of gut microbiota on morbidities in preterm infants. Kaohsiung J Med Sci 2024; 40:780-788. [PMID: 39073226 DOI: 10.1002/kjm2.12878] [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: 06/18/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024] Open
Abstract
The gut microbiota undergoes substantial development from birth, and its development in the initial years of life has a potentially lifelong effect on the health of the individual. However, various factors can disrupt the development of the gut microbiota, leading to a condition known as dysbiosis, particularly in preterm infants. Current studies involving adults have suggested that the gut microbiota not only influences the gut but also has multidimensional effects on remote organs; these pathways are often referred to as the gut-organ axis. Imbalance of the gut microbiota may lead to the development of multiple diseases. Recent studies have revealed that gut dysbiosis in preterm infants may cause several acute morbidities-such as necrotizing enterocolitis, late-onset sepsis, bronchopulmonary dysplasia, and retinopathy of prematurity-and it may also influence long-term outcomes including neurodevelopment and somatic growth. This review mainly presents the existing evidence regarding the relationships between the gut microbiota and these morbidities in preterm infants and explores the role of the gut-organ axis in these morbidities. This paper thus offers insights into the future perspectives on microbiota interventions for promoting the health of preterm infants.
Collapse
Affiliation(s)
- Mei-Yin Lai
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital, School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yin-Hsi Chang
- Department of Ophthalmology, Chang Gung Memorial Hospital, School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chien-Chung Lee
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital, School of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
25
|
Greenwood M, Murciano-Martínez P, Berrington J, Flitsch SL, Austin S, Stewart C. Characterising glycosaminoglycans in human breastmilk and their potential role in infant health. MICROBIAL CELL (GRAZ, AUSTRIA) 2024; 11:221-234. [PMID: 38975022 PMCID: PMC11224681 DOI: 10.15698/mic2024.07.827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 07/09/2024]
Abstract
Human breastmilk is composed of many well researched bioactive components crucial for infant nutrition and priming of the neonatal microbiome and immune system. Understanding these components gives us crucial insight to the health and wellbeing of infants. Research surrounding glycosaminoglycans (GAGs) previously focused on those produced endogenously; however, recent efforts have shifted to understanding GAGs in human breastmilk. The structural complexity of GAGs makes detection and analysis complicated therefore, research is time consuming and limited to highly specialised teams experienced in carbohydrate analysis. In breastmilk, GAGs are present in varying quantities in four forms; chondroitin sulphate, heparin/heparan sulphate, dermatan sulphate and hyaluronic acid, and are hypothesised to behave similar to other bioactive components with suspected roles in pathogen defense and proliferation of beneficial gut bacteria. Chondroitin sulphate and heparin, being the most abundant, are expected to have the most impact on infant health. Their decreasing concentration over lactation further indicates their role and potential importance during early life.
Collapse
Affiliation(s)
- Melissa Greenwood
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle UniversityNewcastle Upon Tyne, NE2 4HHUnited Kingdom
- Analytical Sciences Department, Société des Produits Nestlé, Nestlé Research, Vers-Chez-Les-BlancLausanneSwitzerland
| | - Patricia Murciano-Martínez
- Department of Nutrient Technology, Société des Produits Nestlé, Nestlé Research, Vers-Chez-Les-BlancLausanneSwitzerland
| | - Janet Berrington
- Newcastle Neonatal Service, Royal Victoria Infirmary, Newcastle Upon TyneNE1 4LPUnited Kingdom
| | - Sabine L Flitsch
- School of Chemistry, Faculty of Medical Sciences, The University of Manchester, Manchester Institute of BiotechnologyM1 7DNUnited Kingdom
| | - Sean Austin
- Analytical Sciences Department, Société des Produits Nestlé, Nestlé Research, Vers-Chez-Les-BlancLausanneSwitzerland
| | - Christopher Stewart
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle UniversityNewcastle Upon Tyne, NE2 4HHUnited Kingdom
| |
Collapse
|
26
|
Chen W, Guo K, Huang X, Zhang X, Li X, Chen Z, Wang Y, Wang Z, Liu R, Qiu H, Wang M, Zeng S. The Association of Neonatal Gut Microbiota Community State Types with Birth Weight. CHILDREN (BASEL, SWITZERLAND) 2024; 11:770. [PMID: 39062221 PMCID: PMC11276374 DOI: 10.3390/children11070770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/02/2024] [Accepted: 06/04/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND while most gut microbiota research has focused on term infants, the health outcomes of preterm infants are equally important. Very-low-birth-weight (VLBW) or extremely-low-birth-weight (ELBW) preterm infants have a unique gut microbiota structure, and probiotics have been reported to somewhat accelerate the maturation of the gut microbiota and reduce intestinal inflammation in very-low preterm infants, thereby improving their long-term outcomes. The aim of this study was to investigate the structure of gut microbiota in ELBW neonates to facilitate the early identification of different types of low-birth-weight (LBW) preterm infants. METHODS a total of 98 fecal samples from 39 low-birth-weight preterm infants were included in this study. Three groups were categorized according to different birth weights: ELBW (n = 39), VLBW (n = 39), and LBW (n = 20). The gut microbiota structure of neonates was obtained by 16S rRNA gene sequencing, and microbiome analysis was conducted. The community state type (CST) of the microbiota was predicted, and correlation analysis was conducted with clinical indicators. Differences in the gut microbiota composition among ELBW, VLBW, and LBW were compared. The value of gut microbiota composition in the diagnosis of extremely low birth weight was assessed via a random forest-machine learning approach. RESULTS we briefly analyzed the structure of the gut microbiota of preterm infants with low birth weight and found that the ELBW, VLBW, and LBW groups exhibited gut microbiota with heterogeneous compositions. Low-birth-weight preterm infants showed five CSTs dominated by Enterococcus, Staphylococcus, Klebsiella, Streptococcus, Pseudescherichia, and Acinetobacter. The birth weight and clinical indicators related to prematurity were associated with the CST. We found the composition of the gut microbiota was specific to the different types of low-birth-weight premature infants, namely, ELBW, VLBW, and LBW. The ELBW group exhibited significantly more of the potentially harmful intestinal bacteria Acinetobacter relative to the VLBW and LBW groups, as well as a significantly lower abundance of the intestinal probiotic Bifidobacterium. Based on the gut microbiota's composition and its correlation with low weight, we constructed random forest model classifiers to distinguish ELBW and VLBW/LBW infants. The area under the curve of the classifiers constructed with Enterococcus, Klebsiella, and Acinetobacter was found to reach 0.836 by machine learning evaluation, suggesting that gut microbiota composition may be a potential biomarker for ELBW preterm infants. CONCLUSIONS the gut bacteria of preterm infants showed a CST with Enterococcus, Klebsiella, and Acinetobacter as the dominant genera. ELBW preterm infants exhibit an increase in the abundance of potentially harmful bacteria in the gut and a decrease in beneficial bacteria. These potentially harmful bacteria may be potential biomarkers for ELBW preterm infants.
Collapse
Affiliation(s)
- Wanling Chen
- Shenzhen Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen 518116, China
- Microbiome Therapy Center, South China Hospital, Medical School, Shenzhen University, Shenzhen 518111, China
| | - Kaiping Guo
- Division of Pediatrics, Longgang District Central Hospital of Shenzhen, Shenzhen 518116, China
| | - Xunbin Huang
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Shenzhen 518116, China
| | - Xueli Zhang
- Division of Neonatology, Shenzhen Longhua People’s Hospital, Shenzhen 518109, China
| | - Xiaoxia Li
- Division of Neonatology, Shenzhen Longhua People’s Hospital, Shenzhen 518109, China
| | - Zimiao Chen
- Department of Burn Plastic Surgery, South China Hospital, Shenzhen University, Shenzhen 518111, China
| | - Yanli Wang
- Department of Pediatrics, South China Hospital, Shenzhen University, Shenzhen 518111, China
| | - Zhangxing Wang
- Division of Neonatology, Shenzhen Longhua People’s Hospital, Shenzhen 518109, China
| | - Rongtian Liu
- Department of Pediatrics, Shenzhen Second People’s Hospital, Shenzhen 518035, China
| | - Huixian Qiu
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Shenzhen 518116, China
| | - Mingbang Wang
- Microbiome Therapy Center, South China Hospital, Medical School, Shenzhen University, Shenzhen 518111, China
- Department of Neonatology, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen 518172, China
| | - Shujuan Zeng
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Shenzhen 518116, China
| |
Collapse
|
27
|
Jara J, Alba C, Del Campo R, Fernández L, Sáenz de Pipaón M, Rodríguez JM, Orgaz B. Linking preterm infant gut microbiota to nasograstric enteral feeding tubes: exploring potential interactions and microbial strain transmission. Front Pediatr 2024; 12:1397398. [PMID: 38952433 PMCID: PMC11215057 DOI: 10.3389/fped.2024.1397398] [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: 03/07/2024] [Accepted: 06/03/2024] [Indexed: 07/03/2024] Open
Abstract
Introduction Preterm birth is a growing problem worldwide. Staying at a neonatal intensive care unit (NICU) after birth is critical for the survival of preterm infants whose feeding often requires the use of nasogastric enteral feeding tubes (NEFT). These can be colonized by hospital-associated pathobionts that can access the gut of the preterm infants through this route. Since the gut microbiota is the most impactful factor on maturation of the immune system, any disturbance in this may condition their health. Therefore, the aim of this study is to assess the impact of NEFT-associated microbial communities on the establishment of the gut microbiota in preterm infants. Material and methods A metataxonomic analysis of fecal and NEFT-related samples obtained during the first 2 weeks of life of preterm infants was performed. The potential sharing of strains isolated from the same set of samples of bacterial species involved in NICU's outbreaks, was assessed by Random Amplification of Polymorphic DNA (RAPD) genotyping. Results In the samples taken 48 h after birth (NEFT-1 and Me/F1), Staphylococcus spp. was the most abundant genera (62% and 14%, respectively) and it was latter displaced to 5.5% and 0.45%, respectively by Enterobacteriaceae. Significant differences in beta diversity were detected in NEFT and fecal samples taken at day 17 after birth (NEFT-3 and F3) (p = 0.003 and p = 0.024, respectively). Significant positive correlations were found between the most relevant genera detected in NEFT-3 and F3. 28% of the patients shared at least one RAPD-PCR profile in fecal and NEFT samples and 11% of the total profiles were found at least once simultaneously in NEFT and fecal samples from the same patient. Conclusion The results indicate a parallel bacterial colonization of the gut of preterm neonates and the NEFTs used for feeding, potentially involving strain sharing between these niches. Moreover, the same bacterial RAPD profiles were found in neonates hospitalized in different boxes, suggesting a microbial transference within the NICU environment. This study may assist clinical staff in implementing best practices to mitigate the spread of pathogens that could threaten the health of preterm infants.
Collapse
Affiliation(s)
- J. Jara
- Department of Galenic Pharmacy and Food Science, School of Veterinary Sciences, University Complutense of Madrid (UCM), Madrid, Spain
| | - C. Alba
- Department of Nutrition and Food Science, School of Veterinary Sciences, University Complutense of Madrid (UCM), Madrid, Spain
| | - R. Del Campo
- Department of Microbiology, Ramón y Cajal University Hospital and Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain
| | - L. Fernández
- Department of Galenic Pharmacy and Food Science, School of Veterinary Sciences, University Complutense of Madrid (UCM), Madrid, Spain
| | - M. Sáenz de Pipaón
- Department of Neonatology, La Paz University Hospital of Madrid, Madrid, Spain
- Department of Pediatrics, Autonoma University of Madrid, Madrid, Spain
| | - J. M. Rodríguez
- Department of Nutrition and Food Science, School of Veterinary Sciences, University Complutense of Madrid (UCM), Madrid, Spain
| | - B. Orgaz
- Department of Galenic Pharmacy and Food Science, School of Veterinary Sciences, University Complutense of Madrid (UCM), Madrid, Spain
| |
Collapse
|
28
|
Sánchez-González SG, Cárdenas-del-Castillo BG, Garza-González E, Padilla-Rivas GR, Rodríguez-Balderrama I, Treviño-Garza C, Montes-Tapia FF, Palacios-Saucedo GC, Gutiérrez-Rodríguez A, de-la-O-Cavazos ME. Gut microbiota in preterm infants receiving breast milk or mixed feeding. World J Clin Pediatr 2024; 13:90499. [PMID: 38947995 PMCID: PMC11212766 DOI: 10.5409/wjcp.v13.i2.90499] [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: 12/05/2023] [Revised: 03/27/2024] [Accepted: 04/17/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND Preterm birth is the leading cause of mortality in newborns, with very-low-birth-weight infants usually experiencing several complications. Breast milk is considered the gold standard of nutrition, especially for preterm infants with delayed gut colonization, because it contains beneficial microorganisms, such as Lactobacilli and Bifidobacteria. AIM To analyze the gut microbiota of breastfed preterm infants with a birth weight of 1500 g or less. METHODS An observational study was performed on preterm infants with up to 36.6 wk of gestation and a birth weight of 1500 g or less, born at the University Hospital Dr. José Eleuterio González at Monterrey, Mexico. A total of 40 preterm neonates were classified into breast milk feeding (BM) and mixed feeding (MF) groups (21 in the BM group and 19 in the MF group), from October 2017 to June 2019. Fecal samples were collected before they were introduced to any feeding type. After full enteral feeding was achieved, the composition of the gut microbiota was analyzed using 16S rRNA gene sequencing. Numerical variables were compared using Student's t-test or using the Mann-Whitney U test for nonparametric variables. Dominance, evenness, equitability, Margalef's index, Fisher's alpha, Chao-1 index, and Shannon's diversity index were also calculated. RESULTS No significant differences were observed at the genus level between the groups. Class comparison indicated higher counts of Alphaproteobacteria and Betaproteobacteria in the initial compared to the final sample of the BM group (P < 0.011). In addition, higher counts of Gammaproteobacteria were detected in the final than in the initial sample (P = 0.040). According to the Margalef index, Fisher's alpha, and Chao-1 index, a decrease in species richness from the initial to the final sample, regardless of the feeding type, was observed (P < 0.050). The four predominant phyla were Bacteroidetes, Actinobacteria, Firmicutes, and Proteobacteria, with Proteobacteria being the most abundant. However, no significant differences were observed between the initial and final samples at the phylum level. CONCLUSION Breastfeeding is associated with a decrease in Alphaproteobacteria and Betaproteobacteria and an increase of Gammaproteobacteria, contributing to the literature of the gut microbiota structure of very low-birth-weight, preterm.
Collapse
Affiliation(s)
- Sandra Gabriela Sánchez-González
- Department of Pediatrics, Hospital Universitario “Dr. José Eleuterio González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, México
| | | | - Elvira Garza-González
- Departamento de Bioquímica y Medicina Molecular, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, México
| | - Gerardo R Padilla-Rivas
- Department of Biochemistry and Molecular Medicine, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, México
| | - Isaías Rodríguez-Balderrama
- Department of Pediatrics, Hospital Universitario “Dr. José Eleuterio González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, México
| | - Consuelo Treviño-Garza
- Department of Pediatrics, Hospital Universitario “Dr. José Eleuterio González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, México
| | - Fernando Félix Montes-Tapia
- Department of Pediatric Surgery, Hospital Universitario "Dr. José Eleuterio González", Monterrey 64460, Nuevo León, México
| | - Gerardo C Palacios-Saucedo
- Department of Pediatrics, Hospital Universitario “Dr. José Eleuterio González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, México
| | - Anthony Gutiérrez-Rodríguez
- Department of Pediatrics, Hospital Universitario “Dr. José Eleuterio González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, México
| | - Manuel Enrique de-la-O-Cavazos
- Department of Pediatrics, Hospital Universitario “Dr. José Eleuterio González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, México
| |
Collapse
|
29
|
Lee CC, Chiu CH. Link between gut microbiota and neonatal sepsis. J Formos Med Assoc 2024; 123:638-646. [PMID: 37821302 DOI: 10.1016/j.jfma.2023.09.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 09/15/2023] [Accepted: 09/28/2023] [Indexed: 10/13/2023] Open
Abstract
In neonates, the gastrointestinal tract is rapidly colonized by bacteria after birth. Gut microbiota development is critical during the first few years of life. However, disruption of gut microbiota development in neonates can lead to gut dysbiosis, characterized by overcolonization by pathogenic bacteria and delayed or failed maturation toward increasing microbial diversity and Fermicutes dominance. Gut dysbiosis can predispose infants to sepsis. Pathogenic bacteria can colonize the gut prior to sepsis and cause sepsis through translocation. This review explores gut microbiota development in neonates, the evidence linking gut dysbiosis to neonatal sepsis, and the potential role of probiotics in gut microbiota modulation and sepsis prevention.
Collapse
Affiliation(s)
- Chien-Chung Lee
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Cheng-Hsun Chiu
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan; Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| |
Collapse
|
30
|
Whiteside SA, Odom John AR. Oops, the microbes did it again: Gut dysbiosis precedes late-onset meningitis. J Infect Dis 2024:jiae266. [PMID: 38779890 DOI: 10.1093/infdis/jiae266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024] Open
Affiliation(s)
- Samantha A Whiteside
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Audrey R Odom John
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
31
|
Song F, Li Q, Cui J, Wang J, Xiao S, Yu B, Sun Y, Song W, Wu L, Zhou Y. Exploring the gut microbiota-hippocampus-metabolites axis dysregulation in sepsis mice. Front Microbiol 2024; 15:1302907. [PMID: 38827158 PMCID: PMC11140095 DOI: 10.3389/fmicb.2024.1302907] [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: 12/28/2023] [Accepted: 03/18/2024] [Indexed: 06/04/2024] Open
Abstract
Background Sepsis is commonly associated with a sudden impairment of brain function, thus leading to significant rates of illness and mortality. The objective of this research was to integrate microbiome and metabolome to reveal the mechanism of microbiota-hippocampus-metabolites axis dysfunction in a mouse model of sepsis. Methods A mouse model of sepsis was established via cecal ligation and puncture. The potential associations between the composition of the gut microbiota and metabolites in the hippocampus of mice with sepsis were investigated by combining 16S ribosomal RNA gene sequencing and ultra-high-performance liquid chromatography tandem mass spectrometry. Results A total of 140 differential metabolites were identified in the hippocampal tissues of mice with sepsis when compared to those of control mice. These differential metabolites in mice with sepsis were not only associated with autophagy and serotonergic synapse, but also involved in the metabolism and synthesis of numerous amino acids. At the phylum level, the abundance of Bacteroidota was increased, while that of Firmicutes (Bacillota) was decreased in mice with sepsis. At the genus level, the abundance of Alistipes was increased, while that of Lachnospiraceae_NK4A136_group was decreased in mice with sepsis. The Firmicutes (Bacillota)/Bacteroidota (F/B) ratio was decreased in mice with sepsis when compared to that of control mice. Furthermore, the F/B ratio was positively correlated with 5'-methylthioadenosine, PC (18:3(9Z,12Z,15Z)/18:0) and curdione, and negatively correlated with indoxylsulfuric acid, corticosterone, kynurenine and ornithine. Conclusion Analysis revealed a reduction in the F/B ratio in mice with sepsis, thus contributing to the disturbance of 5'-methylthioadenosine, curdione, PC (18:3(9Z,12Z,15Z)/18:0), corticosterone, ornithine, indoxylsulfuric acid and kynurenine; eventually, these changes led to hippocampus dysfunction. Our findings provide a new direction for the management of sepsis-induced hippocampus dysfunction.
Collapse
Affiliation(s)
- Fangqiang Song
- Department of Critical Care Medicine, Tengzhou Central People’s Hospital, Tengzhou, China
| | - Qinglun Li
- Department of Critical Care Medicine, Tengzhou Central People’s Hospital, Tengzhou, China
| | - Jiyao Cui
- Department of Critical Care Medicine, Tengzhou Central People’s Hospital, Tengzhou, China
| | - Jianhua Wang
- Translational Pharmaceutical Laboratory, Jining NO. 1 People’s Hospital, Jining, China
| | - Shuai Xiao
- Department of Critical Care Medicine, Tengzhou Central People’s Hospital, Tengzhou, China
| | - Bo Yu
- Department of Critical Care Medicine, Tengzhou Central People’s Hospital, Tengzhou, China
| | - Yanqi Sun
- Department of Critical Care Medicine, Tengzhou Central People’s Hospital, Tengzhou, China
| | - Wenke Song
- Department of Critical Care Medicine, Tengzhou Central People’s Hospital, Tengzhou, China
| | - Linlin Wu
- Department of Oncology, Tengzhou Central People’s Hospital, Tengzhou, China
| | - Yongqin Zhou
- Department of Critical Care Medicine, Tengzhou Central People’s Hospital, Tengzhou, China
| |
Collapse
|
32
|
de Kroon RR, Frerichs NM, Struys EA, de Boer NK, de Meij TGJ, Niemarkt HJ. The Potential of Fecal Volatile Organic Compound Analysis for the Early Diagnosis of Late-Onset Sepsis in Preterm Infants: A Narrative Review. SENSORS (BASEL, SWITZERLAND) 2024; 24:3162. [PMID: 38794014 PMCID: PMC11124895 DOI: 10.3390/s24103162] [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: 03/26/2024] [Revised: 05/01/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024]
Abstract
Early diagnosis and treatment of late-onset sepsis (LOS) is crucial for survival, but challenging. Intestinal microbiota and metabolome alterations precede the clinical onset of LOS, and the preterm gut is considered an important source of bacterial pathogens. Fecal volatile organic compounds (VOCs), formed by physiologic and pathophysiologic metabolic processes in the preterm gut, reflect a complex interplay between the human host, the environment, and microbiota. Disease-associated fecal VOCs can be detected with an array of devices with various potential for the development of a point-of-care test (POCT) for preclinical LOS detection. While characteristic VOCs for common LOS pathogens have been described, their VOC profiles often overlap with other pathogens due to similarities in metabolic pathways, hampering the construction of species-specific profiles. Clinical studies have, however, successfully discriminated LOS patients from healthy individuals using fecal VOC analysis with the highest predictive value for Gram-negative pathogens. This review discusses the current advancements in the development of a non-invasive fecal VOC-based POCT for early diagnosis of LOS, which may potentially provide opportunities for early intervention and targeted treatment and could improve clinical neonatal outcomes. Identification of confounding variables impacting VOC synthesis, selection of an optimal detection device, and development of standardized sampling protocols will allow for the development of a novel POCT in the near future.
Collapse
Affiliation(s)
- Rimke R. de Kroon
- Department of Pediatric Gastroenterology, Emma Children’s Hospital, Amsterdam Gastroenterology Endocrinology Metabolism Research Institute, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Nina M. Frerichs
- Department of Pediatric Gastroenterology, Emma Children’s Hospital, Amsterdam Gastroenterology Endocrinology Metabolism Research Institute, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Eduard A. Struys
- Department of Laboratory Medicine, Amsterdam University Medical Center, Location AMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Nanne K. de Boer
- Department of Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism Research Institute, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Tim G. J. de Meij
- Department of Pediatric Gastroenterology, Emma Children’s Hospital, Amsterdam Gastroenterology Endocrinology Metabolism Research Institute, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Hendrik J. Niemarkt
- Department of Neonatology, Maxima Medisch Centrum, De Run 4600, 5504 DB Veldhoven, The Netherlands
- Department of Electrical Engineering, TU Eindhoven, Eindhoven University of Technology, Postbus 513, 5600 MB Eindhoven, The Netherlands
| |
Collapse
|
33
|
Odriozola A, González A, Odriozola I, Álvarez-Herms J, Corbi F. Microbiome-based precision nutrition: Prebiotics, probiotics and postbiotics. ADVANCES IN GENETICS 2024; 111:237-310. [PMID: 38908901 DOI: 10.1016/bs.adgen.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/24/2024]
Abstract
Microorganisms have been used in nutrition and medicine for thousands of years worldwide, long before humanity knew of their existence. It is now known that the gut microbiota plays a key role in regulating inflammatory, metabolic, immune and neurobiological processes. This text discusses the importance of microbiota-based precision nutrition in gut permeability, as well as the main advances and current limitations of traditional probiotics, new-generation probiotics, psychobiotic probiotics with an effect on emotional health, probiotic foods, prebiotics, and postbiotics such as short-chain fatty acids, neurotransmitters and vitamins. The aim is to provide a theoretical context built on current scientific evidence for the practical application of microbiota-based precision nutrition in specific health fields and in improving health, quality of life and physiological performance.
Collapse
Affiliation(s)
- Adrián Odriozola
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain.
| | - Adriana González
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Iñaki Odriozola
- Health Department of Basque Government, Donostia-San Sebastián, Spain
| | - Jesús Álvarez-Herms
- Phymo® Lab, Physiology, and Molecular Laboratory, Collado Hermoso, Segovia, Spain
| | - Francesc Corbi
- Institut Nacional d'Educació Física de Catalunya (INEFC), Centre de Lleida, Universitat de Lleida (UdL), Lleida, Spain
| |
Collapse
|
34
|
Younge N. Influence of infant microbiome on health and development. Clin Exp Pediatr 2024; 67:224-231. [PMID: 37605538 PMCID: PMC11065641 DOI: 10.3345/cep.2023.00598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/08/2023] [Accepted: 07/19/2023] [Indexed: 08/23/2023] Open
Abstract
The microbiome is a complex ecosystem comprising microbes, their genomes, and the surrounding environment. The microbiome plays a critical role in early human development, including maturation of the host immune system and gastrointestinal tract. Multiple factors, including diet, anti-biotic use, and other environmental exposures, influence the establishment of the microbiome during infancy. Numerous studies have identified associations between the microbiome and neonatal diseases, including necrotizing enterocolitis, sepsis, and malnutrition. Furthermore, there is compelling evidence that perturbation of the microbiome in early life can have lasting developmental effects that increase an individual's risk for immune and metabolic diseases in later life. Supplementation of the microbiome with probiotics reduces the risk of necrotizing enterocolitis and sepsis in at-risk infants. This review focuses on the structure and function of the infant microbiome, the environmental and clinical factors that influence its assembly, and its impact on infant health and development.
Collapse
|
35
|
Adeniyi-Ipadeola GO, Hoffman KL, Yang H, Javornik Cregeen SJ, Preidis GA, Ramani S, Hair AB. Human milk cream alters intestinal microbiome of preterm infants: a prospective cohort study. Pediatr Res 2024; 95:1564-1571. [PMID: 38228744 DOI: 10.1038/s41390-023-02948-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 11/09/2023] [Accepted: 11/18/2023] [Indexed: 01/18/2024]
Abstract
BACKGROUND In very low birth weight (VLBW) infants, human milk cream added to standard human milk fortification is used to improve growth. This study aimed to evaluate the impact of cream supplement on the intestinal microbiome of VLBW infants. METHODS Whole genome shotgun sequencing was performed on stool (n = 57) collected from a cohort of 23 infants weighing 500-1250 grams (control = 12, cream = 11). Both groups received an exclusive human milk diet (mother's own milk, donor human milk, and donor human milk-derived fortifier) with the cream group receiving an additional 2 kcal/oz cream at 100 mL/kg/day of fortified feeds and then 4 kcal/oz if poor growth. RESULTS While there were no significant differences in alpha diversity, infants receiving cream significantly differed from infants in the control group in beta diversity. Cream group samples had significantly higher prevalence of Proteobacteria and significantly lower Firmicutes compared to control group. Klebsiella species dominated the microbiota of cream-exposed infants, along with bacterial pathways involved in lipid metabolism and metabolism of cofactors and amino acids. CONCLUSIONS Cream supplementation significantly altered composition of the intestinal microbiome of VLBW infants to favor increased prevalence of Proteobacteria and functional gene content associated with these bacteria. IMPACT We report changes to the intestinal microbiome associated with administration of human milk cream; a novel supplement used to improve growth rates of preterm very low birth weight infants. Since little is known about the impact of cream on intestinal microbiota composition of very low birth weight infants, our study provides valuable insight on the effects of diet on the microbiome of this population. Dietary supplements administered to preterm infants in neonatal intensive care units have the potential to influence the intestinal microbiome composition which may affect overall health status of the infant.
Collapse
Affiliation(s)
- Grace O Adeniyi-Ipadeola
- Graduate Program in Immunology & Microbiology, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Kristi L Hoffman
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Heeju Yang
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | - Sara J Javornik Cregeen
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Geoffrey A Preidis
- Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| | - Sasirekha Ramani
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Amy B Hair
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA.
| |
Collapse
|
36
|
Wala SJ, Ragan MV, Pryor E, Canvasser J, Diefenbach KA, Besner GE. Contemporary use of prophylactic probiotics in NICUs in the United States: a survey update. J Perinatol 2024; 44:739-744. [PMID: 38553600 DOI: 10.1038/s41372-024-01952-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/14/2024] [Accepted: 03/20/2024] [Indexed: 05/15/2024]
Abstract
OBJECTIVE In 2015, 14.0% of US NICUs administered probiotics to very low birth weight infants. Current probiotic use prior to and after the Fall of 2023 (when FDA warnings were issued) remains unknown. STUDY DESIGN A survey was distributed to the American Academy of Pediatrics Section on Neonatal and Perinatal Medicine (August-November/2022) and Neonatology Solutions' Level III/IV NICUs (January-April/2023). Probiotic administration practices were investigated. RESULTS In total, 289 unique NICUs and 406 providers responded to the survey. Of those, 29.1% of NICUs administered prophylactic probiotics to premature neonates, however, this decreased considerably after FDA warnings were issued. Additionally, 71.4% of providers stated willingness to administer probiotics to premature infants if there was an FDA-approved formulation. CONCLUSIONS Probiotic use in US NICUs increased between 2015 and the Fall of 2023 and then dropped dramatically following warning letters from the FDA. The introduction of an FDA-approved probiotic may further expand administration.
Collapse
Affiliation(s)
- Samantha J Wala
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| | - Mecklin V Ragan
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| | | | | | - Karen A Diefenbach
- Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| | - Gail E Besner
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA.
- Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH, USA.
| |
Collapse
|
37
|
Gao Y, Liu L, Cui Y, Zhang J, Wu X. The causality of gut microbiota on onset and progression of sepsis: a bi-directional Mendelian randomization analysis. Front Immunol 2024; 15:1266579. [PMID: 38698853 PMCID: PMC11063379 DOI: 10.3389/fimmu.2024.1266579] [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: 07/25/2023] [Accepted: 03/29/2024] [Indexed: 05/05/2024] Open
Abstract
Background Several observational studies have proposed a potential link between gut microbiota and the onset and progression of sepsis. Nevertheless, the causality of gut microbiota and sepsis remains debatable and warrants more comprehensive exploration. Methods We conducted a two-sample Mendelian randomization (MR) analysis to test the causality between gut microbiota and the onset and progression of sepsis. The genome-wide association study (GWAS) summary statistics for 196 bacterial traits were extracted from the MiBioGen consortium, whereas the GWAS summary statistics for sepsis and sepsis-related outcomes came from the UK Biobank. The inverse-variance weighted (IVW) approach was the primary method used to examine the causal association. To complement the IVW method, we utilized four additional MR methods. We performed a series of sensitivity analyses to examine the robustness of the causal estimates. Results We assessed the causality of 196 bacterial traits on sepsis and sepsis-related outcomes. Genus Coprococcus2 [odds ratio (OR) 0.81, 95% confidence interval (CI) (0.69-0.94), p = 0.007] and genus Dialister (OR 0.85, 95% CI 0.74-0.97, p = 0.016) had a protective effect on sepsis, whereas genus Ruminococcaceae UCG011 (OR 1.10, 95% CI 1.01-1.20, p = 0.024) increased the risk of sepsis. When it came to sepsis requiring critical care, genus Anaerostipes (OR 0.49, 95% CI 0.31-0.76, p = 0.002), genus Coprococcus1 (OR 0.65, 95% CI 0.43-1.00, p = 0.049), and genus Lachnospiraceae UCG004 (OR 0.51, 95% CI 0.34-0.77, p = 0.001) emerged as protective factors. Concerning 28-day mortality of sepsis, genus Coprococcus1 (OR 0.67, 95% CI 0.48-0.94, p = 0.020), genus Coprococcus2 (OR 0.48, 95% CI 0.27-0.86, p = 0.013), genus Lachnospiraceae FCS020 (OR 0.70, 95% CI 0.52-0.95, p = 0.023), and genus Victivallis (OR 0.82, 95% CI 0.68-0.99, p = 0.042) presented a protective effect, whereas genus Ruminococcus torques group (OR 1.53, 95% CI 1.00-2.35, p = 0.049), genus Sellimonas (OR 1.25, 95% CI 1.04-1.50, p = 0.019), and genus Terrisporobacter (OR 1.43, 95% CI 1.02-2.02, p = 0.040) presented a harmful effect. Furthermore, genus Coprococcus1 (OR 0.42, 95% CI 0.19-0.92, p = 0.031), genus Coprococcus2 (OR 0.34, 95% CI 0.14-0.83, p = 0.018), and genus Ruminiclostridium6 (OR 0.43, 95% CI 0.22-0.83, p = 0.012) were associated with a lower 28-day mortality of sepsis requiring critical care. Conclusion This MR analysis unveiled a causality between the 21 bacterial traits and sepsis and sepsis-related outcomes. Our findings may help the development of novel microbiota-based therapeutics to decrease the morbidity and mortality of sepsis.
Collapse
Affiliation(s)
| | | | | | | | - Xiuying Wu
- Department of Anesthesia, ShengJing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
38
|
Williams A. Multiomics data integration, limitations, and prospects to reveal the metabolic activity of the coral holobiont. FEMS Microbiol Ecol 2024; 100:fiae058. [PMID: 38653719 PMCID: PMC11067971 DOI: 10.1093/femsec/fiae058] [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: 09/26/2023] [Revised: 03/25/2024] [Accepted: 04/22/2024] [Indexed: 04/25/2024] Open
Abstract
Since their radiation in the Middle Triassic period ∼240 million years ago, stony corals have survived past climate fluctuations and five mass extinctions. Their long-term survival underscores the inherent resilience of corals, particularly when considering the nutrient-poor marine environments in which they have thrived. However, coral bleaching has emerged as a global threat to coral survival, requiring rapid advancements in coral research to understand holobiont stress responses and allow for interventions before extensive bleaching occurs. This review encompasses the potential, as well as the limits, of multiomics data applications when applied to the coral holobiont. Synopses for how different omics tools have been applied to date and their current restrictions are discussed, in addition to ways these restrictions may be overcome, such as recruiting new technology to studies, utilizing novel bioinformatics approaches, and generally integrating omics data. Lastly, this review presents considerations for the design of holobiont multiomics studies to support lab-to-field advancements of coral stress marker monitoring systems. Although much of the bleaching mechanism has eluded investigation to date, multiomic studies have already produced key findings regarding the holobiont's stress response, and have the potential to advance the field further.
Collapse
Affiliation(s)
- Amanda Williams
- Microbial Biology Graduate Program, Rutgers University, 76 Lipman Drive, New Brunswick, NJ 08901, United States
- Department of Biochemistry and Microbiology, Rutgers University, 76 Lipman Drive, New Brunswick, NJ 08901, United States
| |
Collapse
|
39
|
Patangia DV, Grimaud G, O'Shea CA, Ryan CA, Dempsey E, Stanton C, Ross RP. Early life exposure of infants to benzylpenicillin and gentamicin is associated with a persistent amplification of the gut resistome. MICROBIOME 2024; 12:19. [PMID: 38310316 PMCID: PMC10837951 DOI: 10.1186/s40168-023-01732-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 11/24/2023] [Indexed: 02/05/2024]
Abstract
BACKGROUND Infant gut microbiota is highly malleable, but the long-term longitudinal impact of antibiotic exposure in early life, together with the mode of delivery on infant gut microbiota and resistome, is not extensively studied. METHODS Two hundred and eight samples from 45 infants collected from birth until 2 years of age over five time points (week 1, 4, 8, 24, year 2) were analysed. Based on shotgun metagenomics, the gut microbial composition and resistome profile were compared in the early life of infants divided into three groups: vaginal delivery/no-antibiotic in the first 4 days of life, C-section/no-antibiotic in the first 4 days of life, and C-section/antibiotic exposed in first 4 days of life. Gentamycin and benzylpenicillin were the most commonly administered antibiotics during this cohort's first week of life. RESULTS Newborn gut microbial composition differed in all three groups, with higher diversity and stable composition seen at 2 years of age, compared to week 1. An increase in microbial diversity from week 1 to week 4 only in the C-section/antibiotic-exposed group reflects the effect of antibiotic use in the first 4 days of life, with a gradual increase thereafter. Overall, a relative abundance of Actinobacteria and Bacteroides was significantly higher in vaginal delivery/no-antibiotic while Proteobacteria was higher in C-section/antibiotic-exposed infants. Strains from species belonging to Bifidobacterium and Bacteroidetes were generally persistent colonisers, with Bifidobacterium breve and Bifidobacterium bifidum species being the major persistent colonisers in all three groups. Bacteroides persistence was dominant in the vaginal delivery/no-antibiotic group, with species Bacteroides ovatus and Phocaeicola vulgatus found to be persistent colonisers in the no-antibiotic groups. Most strains carrying antibiotic-resistance genes belonged to phyla Proteobacteria and Firmicutes, with the C-section/antibiotic-exposed group presenting a higher frequency of antibiotic-resistance genes (ARGs). CONCLUSION These data show that antibiotic exposure has an immediate and persistent effect on the gut microbiome in early life. As such, the two antibiotics used in the study selected for strains (mainly Proteobacteria) which were multiple drug-resistant (MDR), presumably a reflection of their evolutionary lineage of historical exposures-leading to what can be an extensive and diverse resistome. Video Abstract.
Collapse
Affiliation(s)
- Dhrati V Patangia
- School of Microbiology, University College Cork, Cork, Ireland
- Teagasc Food Research Centre, Moorepark, Fermoy Co., Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | - Ghjuvan Grimaud
- Teagasc Food Research Centre, Moorepark, Fermoy Co., Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | | | - C A Ryan
- APC Microbiome Ireland, Cork, Ireland
| | - Eugene Dempsey
- APC Microbiome Ireland, Cork, Ireland
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
- Infant Research Centre, University College Cork, Cork, Ireland
| | - Catherine Stanton
- Teagasc Food Research Centre, Moorepark, Fermoy Co., Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | - R Paul Ross
- School of Microbiology, University College Cork, Cork, Ireland.
- APC Microbiome Ireland, Cork, Ireland.
| |
Collapse
|
40
|
Wu J, Singleton SS, Bhuiyan U, Krammer L, Mazumder R. Multi-omics approaches to studying gastrointestinal microbiome in the context of precision medicine and machine learning. Front Mol Biosci 2024; 10:1337373. [PMID: 38313584 PMCID: PMC10834744 DOI: 10.3389/fmolb.2023.1337373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 12/27/2023] [Indexed: 02/06/2024] Open
Abstract
The human gastrointestinal (gut) microbiome plays a critical role in maintaining host health and has been increasingly recognized as an important factor in precision medicine. High-throughput sequencing technologies have revolutionized -omics data generation, facilitating the characterization of the human gut microbiome with exceptional resolution. The analysis of various -omics data, including metatranscriptomics, metagenomics, glycomics, and metabolomics, holds potential for personalized therapies by revealing information about functional genes, microbial composition, glycans, and metabolites. This multi-omics approach has not only provided insights into the role of the gut microbiome in various diseases but has also facilitated the identification of microbial biomarkers for diagnosis, prognosis, and treatment. Machine learning algorithms have emerged as powerful tools for extracting meaningful insights from complex datasets, and more recently have been applied to metagenomics data via efficiently identifying microbial signatures, predicting disease states, and determining potential therapeutic targets. Despite these rapid advancements, several challenges remain, such as key knowledge gaps, algorithm selection, and bioinformatics software parametrization. In this mini-review, our primary focus is metagenomics, while recognizing that other -omics can enhance our understanding of the functional diversity of organisms and how they interact with the host. We aim to explore the current intersection of multi-omics, precision medicine, and machine learning in advancing our understanding of the gut microbiome. A multidisciplinary approach holds promise for improving patient outcomes in the era of precision medicine, as we unravel the intricate interactions between the microbiome and human health.
Collapse
Affiliation(s)
- Jingyue Wu
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
| | - Stephanie S. Singleton
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
| | - Urnisha Bhuiyan
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
| | - Lori Krammer
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
- Milken Institute School of Public Health, The George Washington University, Washington, DC, United States
| | - Raja Mazumder
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
- The McCormick Genomic and Proteomic Center, The George Washington University, Washington, DC, United States
| |
Collapse
|
41
|
VALENTINE GREGORYC, WALLEN LINDAD. Neonatal Bacterial Sepsis and Meningitis. AVERY'S DISEASES OF THE NEWBORN 2024:439-449.e5. [DOI: 10.1016/b978-0-323-82823-9.00033-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
42
|
Chang HY, Lin CY, Chiang Chiau JS, Chang JH, Hsu CH, Ko MHJ, Lee HC. Probiotic supplementation modifies the gut microbiota profile of very low birth weight preterm infants during hospitalization. Pediatr Neonatol 2024; 65:55-63. [PMID: 37500417 DOI: 10.1016/j.pedneo.2023.06.002] [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: 03/16/2023] [Revised: 05/09/2023] [Accepted: 06/07/2023] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND Probiotic supplementation is increasingly being given to very low birth weight (VLBW) preterm infants. This preliminary observational study aimed to investigate the effects of multiple-strain probiotics on the gut microbiota of VLBW preterm infants. METHODS We collected meconium and stool samples on days 14, 30, and 60 after birth from 49 VLBW infants with a gestational age of <32 weeks. The infants were divided into the probiotics (n = 24) and control (n = 25) groups. The microbial composition and diversity in the gut of the two groups were analyzed using 16 S rRNA gene sequencing. RESULTS The relative abundance of Bifidobacterium and Lactobacillus was significantly higher in the probiotics group than in the control group on days 14, 30, and 60 (Bifidobacterium: p = 0.002, p < 0.0001, and p < 0.0001, respectively; Lactobacillus: p = 0.012, p < 0.0001, and p < 0.0001, respectively). The control group exhibited a significantly higher proportion of participants with a low abundance (<1%) of Bifidobacterium or Lactobacillus on days 14, 30, and 60 than those in the probiotic group. Moreover, the probiotics group exhibited a significantly lower abundance of Klebsiella on days 14 and 30 (2.4% vs. 11.6%, p = 0.037; and 7.9% vs. 16.6%, p = 0.032, respectively) and of Escherichia-Shigella on day 60 than the control group (6.1% vs. 12.3%, p = 0.013). Beta diversity analysis revealed that the microbiota profile was clearly divided into two groups on days 30 and 60 (p = 0.001). CONCLUSION Probiotic supplementation significantly increased the relative abundance of Bifidobacterium and Lactobacillus and inhibited the growth of potential pathogens. Furthermore, probiotic supplementation led to a distinct gut microbiota profile. Further research is needed to identify probiotic strains that exert significant influence on the gut microbiome and their long-term health implications in preterm infants.
Collapse
Affiliation(s)
- Hung-Yang Chang
- Department of Pediatrics, MacKay Children's Hospital, Taipei 104, Taiwan; Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan
| | - Chia-Ying Lin
- Department of Pediatrics, MacKay Children's Hospital, Taipei 104, Taiwan
| | | | - Jui-Hsing Chang
- Department of Pediatrics, MacKay Children's Hospital, Taipei 104, Taiwan; Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan
| | - Chyong-Hsin Hsu
- Department of Pediatrics, MacKay Children's Hospital, Taipei 104, Taiwan
| | - Mary Hsin-Ju Ko
- Department of Pediatrics, Hsinchu MacKay Memorial Hospital, Hsinchu 300, Taiwan
| | - Hung-Chang Lee
- Department of Pediatrics, MacKay Children's Hospital, Taipei 104, Taiwan; Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan.
| |
Collapse
|
43
|
Yew WC, Young GR, Nelson A, Cheung W, Stewart CJ, Bridge SH, Granger C, Berrington JE, Embleton ND, Smith DL. The core phageome and its interrelationship with preterm human milk lipids. Cell Rep 2023; 42:113373. [PMID: 37967008 DOI: 10.1016/j.celrep.2023.113373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 08/09/2023] [Accepted: 10/18/2023] [Indexed: 11/17/2023] Open
Abstract
Phages and lipids in human milk (HM) may benefit preterm infant health by preventing gastrointestinal pathobiont overgrowth and microbiome modulation. Lipid association may promote vertical transmission of phages to the infant. Despite this, interrelationships between lipids and phages are poorly characterized in preterm HM. Shotgun metagenomics and untargeted lipidomics of phage and lipid profiles from 99 preterm HM samples reveals that phages are abundant and prevalent from the first week and throughout the first 100 days of lactation. Phage-host richness of preterm HM increases longitudinally. Core phage communities characterized by Staphylococcus- and Propionibacterium-infecting phages are significantly correlated with long-chain fatty acid abundances over lactational age. We report here a phage-lipid interaction in preterm HM, highlighting the potential importance of phage carriage in preterm HM. These results reveal possible strategies for phage carriage in HM and their importance in early-life microbiota development.
Collapse
Affiliation(s)
- Wen C Yew
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne NE1 8ST, UK
| | - Gregory R Young
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne NE1 8ST, UK; Hub for Biotechnology in the Built Environment, Northumbria University, Newcastle upon Tyne NE1 8ST, UK
| | - Andrew Nelson
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne NE1 8ST, UK
| | - William Cheung
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne NE1 8ST, UK
| | - Christopher J Stewart
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Simon H Bridge
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne NE1 8ST, UK; Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Claire Granger
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; Neonatal Medicine, Newcastle upon Tyne Hospitals National Health Service Foundation Trust, Newcastle upon Tyne NE7 7DN, UK
| | - Janet E Berrington
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; Neonatal Medicine, Newcastle upon Tyne Hospitals National Health Service Foundation Trust, Newcastle upon Tyne NE7 7DN, UK
| | - Nicholas D Embleton
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Darren L Smith
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne NE1 8ST, UK; Hub for Biotechnology in the Built Environment, Northumbria University, Newcastle upon Tyne NE1 8ST, UK.
| |
Collapse
|
44
|
Heston SM, Lim CSE, Ong C, Chua MC, Kelly MS, Yeo KT. Strain-resolved metagenomic analysis of the gut as a reservoir for bloodstream infection pathogens among premature infants in Singapore. Gut Pathog 2023; 15:55. [PMID: 37974294 PMCID: PMC10652614 DOI: 10.1186/s13099-023-00583-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Gut dysbiosis contributes to the high risk of bloodstream infection (BSI) among premature infants. Most prior studies of the premature infant gut microbiota were conducted in Western countries and prior to development of current tools for strain-resolved analysis. METHODS We performed metagenomic sequencing of weekly fecal samples from 75 premature infants at a single hospital in Singapore. We evaluated associations between clinical factors and gut microbiota composition using PERMANOVA and mixed effects linear regression. We used inStrain to perform strain-level analyses evaluating for gut colonization by BSI-causing strains. RESULTS Median (interquartile range) gestation was 27 (25, 29) weeks, and 63% of infants were born via Cesarean section. Antibiotic exposures (PERMANOVA; R2 = 0.017, p = 0.001) and postnatal age (R2 = 0.015, p = 0.001) accounted for the largest amount of variability in gut microbiota composition. Increasing postnatal age was associated with higher relative abundances of several common pathogens (Enterococcus faecalis: p < 0.0001; Escherichia coli: p < 0.0001; Klebsiella aerogenes: p < 0.0001; Klebsiella pneumoniae: p < 0.0001). Antibiotic exposures were generally associated with lower relative abundances of both frequently beneficial bacteria (e.g., Bifidobacterium species) and common enteric pathogens (e.g., Enterobacter, Klebsiella species). We identified strains identical to the blood culture isolate in fecal samples from 12 of 16 (75%) infants who developed BSI, including all infections caused by typical enteric bacteria. CONCLUSIONS Antibiotic exposures were the dominant modifiable factor affecting gut microbiota composition in a large cohort of premature infants from South-East Asia. Strain-resolved analyses indicate that the gut is an important reservoir for organisms causing BSI among premature infants.
Collapse
Affiliation(s)
- Sarah M Heston
- Division of Pediatric Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
| | - Charis Shu En Lim
- Department of Neonatology, KK Women's and Children's Hospital, Singapore, Singapore
| | - Chengsi Ong
- Department of Neonatology, KK Women's and Children's Hospital, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
- Department of Nutrition and Dietetics, KK Women's and Children's Hospital, Singapore, Singapore
| | - Mei Chien Chua
- Department of Neonatology, KK Women's and Children's Hospital, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Matthew S Kelly
- Division of Pediatric Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
| | - Kee Thai Yeo
- Department of Neonatology, KK Women's and Children's Hospital, Singapore, Singapore.
- Duke-NUS Medical School, Singapore, Singapore.
| |
Collapse
|
45
|
Wang N, Zhang J, Yu Z, Yan X, Zhang L, Peng H, Chen C, Li R. Oropharyngeal administration of colostrum targeting gut microbiota and metabolites in very preterm infants: protocol for a multicenter randomized controlled trial. BMC Pediatr 2023; 23:508. [PMID: 37845612 PMCID: PMC10577906 DOI: 10.1186/s12887-023-04346-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/03/2023] [Indexed: 10/18/2023] Open
Abstract
BACKGROUND Oropharyngeal administration of colostrum (OAC) has an immune-stimulating effect on oropharyngeal-associated lymphoid tissue, and can promote the maturation of the gastrointestinal tract. However, how OAC promotes intestinal maturation in preterm infants by altering gut microbiota remains unclear. We aim to assess changes in gut microbiota and metabolites after OAC in very preterm infants. METHODS A multicenter, double-blind, randomized controlled trial will be conducted in three large neonatal intensive care units in Shenzhen, China, with preterm infants with gestational age less than 32 weeks at birth and birth weight less than 1500 g. It is estimated that 320 preterm infants will be enrolled in this study within one year. The intervention group will receive oropharyngeal administration of 0.2 ml colostrum every 3 h, starting between the first 48 to 72 h and continued for 5 consecutive days. Following a similar administration scheme, the control group will receive oropharyngeal administration of sterile water. Stool samples will be collected at the first defecation, as well as on the 7th, 14th, 21st and 28th days after birth for analysis of effect of OAC on gut microbiota and metabolites through 16sRNA gene sequencing and liquid chromatography-mass spectrometry. DISCUSSION This proposal advocates for the promotion of OAC as a safe and relatively beneficial protocol in neonatal intensive care units, which may contribute to the establishment of a dominant intestinal flora. Findings of this study may help improve the health outcomes of preterm infants by establishment of targeted gut microbiota in future studies. TRIAL REGISTRATION NCT05481866 (registered July 30, 2022 on ClinicalTrials.gov).
Collapse
Affiliation(s)
- Na Wang
- Department of Neonatology, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Jiangsu, China
| | - Jia Zhang
- Department of Neonatology, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Jiangsu, China
| | - Zhangbin Yu
- Department of Neonatology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China.
| | - Xudong Yan
- Department of Neonatology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Lian Zhang
- Department of Neonatology, Bao'an Maternal and Child Health Hospital, Shenzhen, Guangdong, China
| | - Haibo Peng
- Department of Neonatology, Bao'an Maternal and Child Health Hospital, Shenzhen, Guangdong, China
| | - Cheng Chen
- Department of Neonatology, Longgang Maternal and Child Health Hospital, Shenzhen, Guangdong, China
| | - Rui Li
- Department of Neonatology, Longgang Maternal and Child Health Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
46
|
Modrzejewska M, Bosy-Gąsior W, Grzesiak W. Association of Positive Bacterial Cultures Obtained from the Throat, Anus, Ear, Bronchi and Blood in Very-Low-Birth-Weight Premature Infants with Severe Retinopathy of Prematurity-Own Observations. J Clin Med 2023; 12:6374. [PMID: 37835018 PMCID: PMC10573982 DOI: 10.3390/jcm12196374] [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: 08/02/2023] [Revised: 09/27/2023] [Accepted: 10/03/2023] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND The causative factors responsible for the development of Retinopathy of Prematurity (ROP) are still unexplored. Therefore, one of the most important factors can be perinatal inflammation. METHODS This retrospective study included 114 premature infants (228 eyes) meeting a birth criteria of ≤ 32 weeks gestational age (GA) and a birth weight (BW) ≤ 1710. Examined Group (EG) n = 51 of BW 852.7 ± 255.7; GA 26.3 ± 2.0 with severe ROP treated by diode laser or anti-VEGF intravitreal injection. Control Group (CG) n = 63 of BW 1313.9 ± 284.5; GA 28.8 ± 1.6 without ROP. Microbiological bacterial and fungal cultures of the ear, anus, bronchial throat and blood were taken. Medical data and laboratory tests in correlation to 3 ROP and A-ROP were analysed. RESULTS Positive bacterial tests dominated in EG, 47% vs. CG, 23%. Significant correlations between positive cultures obtained from natural cavities: anus (p < 0.001), throat (p = 0.002), as well as from blood (p = 0.001) and severe ROP which requires diode laser and anti-VEGF treatment were noted. Significant inflammation markers which correlate with the development of severe ROP are Klebsiella pneumoniae (KP) (p = 0.002) and Coagulase-negative Staphylococci (CoNS) (p < 0.001). CoNS, p < 0.001; KP, p = 0.002; the remaining Maltophilia stenotrophomonas (MS); Staphylococcus aureus (SA), p = 0.005; and Enterobacter cloacae (EC), p = 0.02 were the most frequent bacteria in severe ROP. High levels of white blood cells (WBC), C-reactive protein (CRP), lymphocytes (LYM) and low thrombocytes (PLT) correlated sequentially with (Odds Ratio, OR) CoNS (2.3); MS (5.9); KP (3.1); and all positive cultures (APC) (9.5). An important correlation between the BPD-EC (4.3); intrauterine inflammation-KP (3.4); PDA-EC (3.9); and asphyxia-CoNS (3.0) was identified. CONCLUSIONS It cannot be ruled out that positive microbiological results of blood, anal and pharyngeal cultures may become prognostic markers for the early development of ROP, which would enable early initiation of ophthalmological treatment in premature infants from the VLBW group.
Collapse
Affiliation(s)
- Monika Modrzejewska
- Scientific Association of Students, II Department of Ophthalmology, Pomeranian Medical University, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland;
| | - Wiktoria Bosy-Gąsior
- Scientific Association of Students, II Department of Ophthalmology, Pomeranian Medical University, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland;
| | - Wilhelm Grzesiak
- Faculty of Biotechnology and Animal Hysbandry, West-Pomeranian Technological University, Al. Piastów 48, 70-311 Szczecin, Poland;
| |
Collapse
|
47
|
Young GR, Nelson A, Stewart CJ, Smith DL. Bacteriophage communities are a reservoir of unexplored microbial diversity in neonatal health and disease. Curr Opin Microbiol 2023; 75:102379. [PMID: 37647765 DOI: 10.1016/j.mib.2023.102379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 07/30/2023] [Accepted: 08/02/2023] [Indexed: 09/01/2023]
Abstract
Acquisition and development of the gut microbiome are vital for immune education in neonates, especially those born preterm. As such, microbial communities have been extensively studied in the context of postnatal health and disease. Bacterial communities have been the focus of research in this area due to the relative ease of targeted bacterial sequencing and the availability of databases to align and validate sequencing data. Recent increases in high-throughput metagenomic sequencing accessibility have facilitated research to investigate bacteriophages within the context of neonatal gut microbial communities. Focusing on unexplored viral diversity, has identified novel bacteriophage species and previously uncharacterised viral diversity. In doing so, studies have highlighted links between bacteriophages and bacterial community structure in the context of health and disease. However, much remains unknown about the complex relationships between bacteriophages, the bacteria they infect and their human host. With a particular focus on preterm infants, this review highlights opportunities to explore the influence of bacteriophages on developing microbial communities and the tripartite relationships between bacteriophages, bacteria and the neonatal human host. We suggest a focus on expanding collections of isolated bacteriophages that will further our understanding of the growing numbers of bacteriophages identified in metagenomes.
Collapse
Affiliation(s)
- Gregory R Young
- Applied Sciences, Health and Life Sciences, Northumbria University, Newcastle, UK
| | - Andrew Nelson
- Applied Sciences, Health and Life Sciences, Northumbria University, Newcastle, UK
| | | | - Darren L Smith
- Applied Sciences, Health and Life Sciences, Northumbria University, Newcastle, UK.
| |
Collapse
|
48
|
Zhang C, Zhao X, Zhu Z, Wang K, Moon BF, Zhang B, Sadat SN, Guo J, Bao J, Zhang D, Zhang X. Evaluation of white matter microstructural alterations in premature infants with necrotizing enterocolitis. Quant Imaging Med Surg 2023; 13:6412-6423. [PMID: 37869353 PMCID: PMC10585499 DOI: 10.21037/qims-22-195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 08/16/2023] [Indexed: 10/24/2023]
Abstract
Background Preterm infants with necrotizing enterocolitis (NEC) are at high risk of adverse neurodevelopmental outcomes. The aim of this study was to explore the value of diffusion tensor imaging (DTI) combined with serum C-reactive protein (CRP) and procalcitonin (PCT) in evaluating alterations of white matter (WM) microstructure in preterm infants with NEC. Methods A retrospective cross-sectional study was conducted in which all participants were consecutively enrolled at The Third Affiliated Hospital of Zhengzhou University from June 2017 and October 2021. Data from 30 preterm infants with NEC [mean gestational age at birth 31.41±1.15 weeks; mean age at magnetic resonance imaging (MRI) 37.53±3.08 weeks] and 40 healthy preterm infants with no NEC were recorded (mean gestational age at birth 32.27±2.09 weeks; mean age at MRI 37.15±3.23 weeks). WM was used to obtain the fractional anisotropy (FA) and mean diffusivity (MD) values of the regions of interest (ROIs). Additionally, serum levels of CRP and PCT were determined. Spearman correlation analysis was performed between the WM-derived parameters, CRP level, and the PCT serum index. Results Preterm infants with NEC had reduced FA values and elevated MD values in WM regions [posterior limbs of the internal capsule (PLIC), lentiform nucleus (LN), frontal white matter (FWM)] compared to the control group (P<0.05). Additionally, the FA of the PLIC was negatively correlated with serum CRP (r=-0.846; P<0.05) and PCT (r=-0.843; P<0.05). Meanwhile, the MD of PLIC was positively correlated with serum CRP (r=0.743; P<0.05) and PCT (r=0.743; P<0.05, respectively). The area under the curve (AUC) of FA and MD combined with CRP and PCT in the diagnosis of WM microstructure alterations with NEC was 0.968, representing a considerable improvement in predicted efficacy over single indicators, including FA [AUC: 0.938; 95% confidence interval (CI): 0.840-0.950], MD (AUC: 0.807; 95% CI: 0.722-0.838), CRP (AUC: 0.867; 95% CI: 0.822-0.889), and PCT (AUC: 0.706; 95% CI: 0.701-0.758). Conclusions WM can noninvasively and quantitatively assess the WM microstructure alterations in preterm infants with NEC. WM combined with serum CRP and PCT demonstrated superior performance in detecting and evaluating WM microstructure alterations in preterm infants with NEC.
Collapse
Affiliation(s)
- Chunxiang Zhang
- Department of Radiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
| | - Xin Zhao
- Department of Radiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
| | - Zitao Zhu
- Medical College, Wuhan University, Wuhan, China
| | - Kaiyu Wang
- GE HealthCare, MR Research China, Beijing, China
| | - Brianna F. Moon
- Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Bohao Zhang
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
| | | | - Jinxia Guo
- GE HealthCare, MR Research China, Beijing, China
| | - Jieaoxue Bao
- Department of Radiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
| | - Ding Zhang
- Department of Radiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
| | - Xiaoan Zhang
- Department of Radiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
49
|
Zhong W, Danielsson H, Brusselaers N, Wackernagel D, Sjöbom U, Sävman K, Hansen Pupp I, Ley D, Nilsson AK, Fagerberg L, Uhlén M, Hellström A. The development of blood protein profiles in extremely preterm infants follows a stereotypic evolution pattern. COMMUNICATIONS MEDICINE 2023; 3:107. [PMID: 37532738 PMCID: PMC10397184 DOI: 10.1038/s43856-023-00338-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 07/25/2023] [Indexed: 08/04/2023] Open
Abstract
BACKGROUND Preterm birth is the leading cause of neonatal mortality and morbidity. Early diagnosis and interventions are critical to improving the clinical outcomes of extremely premature infants. Blood protein profiling during the first months of life in preterm infants can shed light on the role of early extrauterine development and provide an increased understanding of maturation after extremely preterm birth and the underlying mechanisms of prematurity-related disorders. METHODS We have investigated the blood protein profiles during the first months of life in preterm infants on the role of early extrauterine development. The blood protein levels were analyzed using next generation blood profiling on 1335 serum samples, collected longitudinally at nine time points from birth to full-term from 182 extremely preterm infants. RESULTS The protein analysis reveals evident predestined serum evolution patterns common for all included infants. The majority of the variations in blood protein expression are associated with the postnatal age of the preterm infants rather than any other factors. There is a uniform protein pattern on postnatal day 1 and after 30 weeks postmenstrual age (PMA), independent of gestational age (GA). However, during the first month of life, GA had a significant impact on protein variability. CONCLUSIONS The unified pattern of protein development for all included infants suggests an age-dependent stereotypic development of blood proteins after birth. This knowledge should be considered in neonatal settings and might alter the clinical approach within neonatology, where PMA is today the most dominant age variable.
Collapse
Affiliation(s)
- Wen Zhong
- Science for Life Laboratory, Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, Sweden
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Hanna Danielsson
- Centre for Translational Microbiome Research, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Sach's Children's and Youth Hospital, Södersjukhuset, Stockholm, Sweden
| | - Nele Brusselaers
- Centre for Translational Microbiome Research, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Global Health Institute, Antwerp University, Antwerp, Belgium
| | - Dirk Wackernagel
- Department of Neonatology, Karolinska University Hospital and Institute, Astrid Lindgrens Children's Hospital, Stockholm, Sweden
| | - Ulrika Sjöbom
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Learning and Leadership for Health Care Professionals At the Institute of Health and Care Science at Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Karin Sävman
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Dept of Neonatology, The Queen Silvia Children's Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ingrid Hansen Pupp
- Department of Pediatrics, Institute of Clinical Sciences Lund, Lund University and Skane University Hospital, Lund, Sweden
| | - David Ley
- Department of Pediatrics, Institute of Clinical Sciences Lund, Lund University and Skane University Hospital, Lund, Sweden
| | - Anders K Nilsson
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Linn Fagerberg
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Mathias Uhlén
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Ann Hellström
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
50
|
Schim van der Loeff I, Tsilifis C, Abdelhafiz K, Williams EJ. Recurrent group B stretococcus infection in an extremely premature infant: as a preterm neonate, infant and toddler. BMJ Case Rep 2023; 16:e255216. [PMID: 37507127 PMCID: PMC10387633 DOI: 10.1136/bcr-2023-255216] [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] [Indexed: 07/30/2023] Open
Abstract
We report five discrete episodes of group B streptococcus (GBS) bacteraemia in an extremely premature infant, extending into early childhood. The first four episodes occurred during infancy despite appropriate treatment. Breastmilk was positive for group B streptococcal 16S DNA by polymerase chain reaction. The fifth episode occurred at 17 months of age, shortly after stopping antimicrobial prophylaxis.Radiological investigations did not identify a focus for recurrence of GBS bacteraemia, and immunological investigations and targeted whole genome sequencing yielded only transient hypogammaglobulinaemia of infancy, which resolved.This case highlights invasive GBS infection as a cause of infant morbidity. Premature infants are at particular risk of invasive as well as recurrent disease. GBS is typically a sensitive organism and each episode of GBS in our patient was effectively treated with penicillin. The role of breastmilk in recurrent GBS is controversial; in this case infant and mother isolated identical GBS serotypes and were concurrently treated with rifampicin.
Collapse
Affiliation(s)
- Ina Schim van der Loeff
- Paediatric Immunology and Infectious Diseases, Great North Children's Hospital, Newcastle upon Tyne, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Christo Tsilifis
- Paediatric Immunology and Infectious Diseases, Great North Children's Hospital, Newcastle upon Tyne, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Khalid Abdelhafiz
- Neonatalogy, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Eleri J Williams
- Paediatric Immunology and Infectious Diseases, Great North Children's Hospital, Newcastle upon Tyne, UK
| |
Collapse
|