1
|
Jiang Y, Yang J, Liu Y, Liu Y, Zhou W, Yang B, Hu B, Wu H. A neonate with meningitis caused by probiotic-related Clostridium butyricum. Int J Infect Dis 2025; 155:107900. [PMID: 40185197 DOI: 10.1016/j.ijid.2025.107900] [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: 01/19/2025] [Revised: 03/19/2025] [Accepted: 03/29/2025] [Indexed: 04/07/2025] Open
Abstract
OBJECTIVES Clostridium butyricum (C. butyricum), a normal gut bacterium in humans, is commonly used as a probiotic. We described a 26-day-old premature neonate who was diagnosed with probiotic-related C. butyricum meningitis. DESIGN Upon the admission, suppurative meningitis was considered based on cerebrospinal fluid (CSF) biochemical test and neuroimaging examination, and C. butyricum was subsequently identified by CSF metagenomic next-generation sequencing. Given the history of administrating living C. butyricum products before admission, probiotics-associated suppurative meningitis was considered a high possibility, leading to the confirmation of anti-infection treatment including vancomycin and meropenem. RESULTS Following this therapy, the infant's CSF profiles demonstrated improvement. Additionally, further phylogenetic analysis confirmed the high homologous of C. butyricum from CSF with probiotics. CONCLUSIONS This is the first report of C. butyricum infection in neonates, highlighting the importance for prudence in administrating probiotics for neonates, particularly in high-risk groups such as preterm infants, those with central venous catheters and intestinal diseases.
Collapse
Affiliation(s)
- Yanfang Jiang
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Jiaxu Yang
- Department of Neonatology, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Yong Liu
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Ying Liu
- Department of Neonatology, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Wenli Zhou
- Department of Neonatology, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Bin Yang
- Center for Infectious Diseases, Vision Medicals Co., Ltd, Guangzhou, Guangdong Province, China
| | - Bingxue Hu
- Center for Infectious Diseases, Vision Medicals Co., Ltd, Guangzhou, Guangdong Province, China
| | - Hui Wu
- Department of Neonatology, The First Hospital of Jilin University, Changchun, Jilin Province, China.
| |
Collapse
|
2
|
Han J, Ren Y, Zhang P, Fang C, Yang L, Zhou S, Ji Z. The effectiveness of treatment with probiotics in preventing necrotizing enterocolitis and related mortality: results from an umbrella meta-analysis on meta-analyses of randomized controlled trials. BMC Gastroenterol 2025; 25:245. [PMID: 40217146 PMCID: PMC11987312 DOI: 10.1186/s12876-025-03788-0] [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: 10/28/2024] [Accepted: 03/17/2025] [Indexed: 04/15/2025] Open
Abstract
INTRODUCTION Probiotic supplementation has been proposed as a preventive measure for necrotizing enterocolitis (NEC) in preterm infants. This umbrella meta-analysis assesses the effects of probiotics, including single-strain and multi-strain formulations, on NEC and related mortality. METHODS A comprehensive search was conducted in PubMed, Scopus, ISI Web of Science, and Embase for studies up to August 2024. The AMSTAR2 tool assessed the quality of included studies. Meta-analysis studies were selected based on the PICOS framework, focusing on preterm neonates (< 37-week gestation), probiotic supplementation (single-strain or multi-strain), placebo or standard care comparison, and outcomes of NEC and mortality. Pooled relative risks (RR) and odds ratios (OR) with 95% confidence intervals (CI) were calculated using random-effects models. RESULTS Overall, 35 eligible studies were included into the study. Twenty-six and 32 probiotic intervention arms used single- and multi-strain probiotics, respectively. The findings revealed that probiotics decreased NEC significantly (ESRR: 0.51; 95% CI: 0.46, 0.55, p < 0.001, and ESOR: 0.59; 95%CI: 0.48, 0.72, P < 0.001), and mortality rate (ESRR: 0.72; 95% CI: 0.68, 0.76, P < 0.001, and ESOR: 0.77; 95%CI: 0.70, 0.84, p < 0.001). CONCLUSION The present review suggests that supplementation with probiotics reduced NEC and related mortality. Probiotic supplementation can be recognized as a NEC-preventing approach in preterm and very preterm infants, particularly Multi-strain probiotics.
Collapse
Affiliation(s)
- Jiaju Han
- Department of Gastrointestinal Surgery, Taizhou Hospital, Wenzhou Medical University, No.105 Westgate Street, Linhai, 317000, China
| | - Yufeng Ren
- Department of Gastrointestinal Surgery, Taizhou Hospital, Wenzhou Medical University, No.105 Westgate Street, Linhai, 317000, China.
| | - Peini Zhang
- Department of Gastrointestinal Surgery, Taizhou Hospital, Wenzhou Medical University, No.105 Westgate Street, Linhai, 317000, China
| | - Chengfeng Fang
- Department of Gastrointestinal Surgery, Taizhou Hospital, Wenzhou Medical University, No.105 Westgate Street, Linhai, 317000, China
| | - Leilei Yang
- Department of Gastrointestinal Surgery, Taizhou Hospital, Wenzhou Medical University, No.105 Westgate Street, Linhai, 317000, China
| | - Shenkang Zhou
- Department of Gastrointestinal Surgery, Taizhou Hospital, Wenzhou Medical University, No.105 Westgate Street, Linhai, 317000, China
| | - Zhiqing Ji
- Department of Gastrointestinal Surgery, Taizhou Hospital, Wenzhou Medical University, No.105 Westgate Street, Linhai, 317000, China
| |
Collapse
|
3
|
Menegolla MP, Silveira RC, Görgen ARH, Gandolfi FE, Panceri C, Procianoy RS. Empiric Antibiotic Therapy and Neurodevelopment Outcome of Very Low Birth Weight Infants. Neuropediatrics 2025. [PMID: 40107306 DOI: 10.1055/a-2561-8208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
To determine the effects of empiric antibiotic therapy within the first 72 hours after birth, in cases of suspected early-onset sepsis without positive blood cultures, on the neurodevelopment of VLBW infants.Cohort study conducted from January 2014 to December 2021, included neonates from 24 to 32 weeks' gestation. They were categorized based on receiving early antibiotics. Outcomes measured included neonatal morbidities and scores on the Bayley Scales of Infant Development, Third Edition (BSID-III), at 12 to 36 months corrected age.Of 261 VLBW infants 52.9% (n = 138) received empiric antibiotics within the first 72 hours, while 47.1% (n = 123) did not. Multivariate analysis revealed no association between early antibiotics and neurodevelopmental delay. Severe intraventricular hemorrhage independently correlated with delays, while late-onset sepsis and bronchopulmonary dysplasia contributed to specific motor and cognitive delays. Propensity score matching (PSM) was conducted using various models that included gestational age, late-onset sepsis, severe intraventricular hemorrhage, bronchopulmonary dysplasia, and clinical chorioamnionitis. However, antibiotic use was not independently associated with an increased risk of developmental delay in the applied models.Although the use of antibiotics did not emerge as an independent factor contributing to developmental delay, VLBW infants who received antibiotics had more morbidities during their NICU stay.
Collapse
Affiliation(s)
- Marina P Menegolla
- Newborn Section, Child Health and Adolescent Medicine Graduate Program, Department of Pediatrics, Universidade Federal do Rio Grande do Sul, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Rita C Silveira
- Newborn Section, Child Health and Adolescent Medicine Graduate Program, Department of Pediatrics, Universidade Federal do Rio Grande do Sul, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Antônio R H Görgen
- Newborn Section, Child Health and Adolescent Medicine Graduate Program, Department of Pediatrics, Universidade Federal do Rio Grande do Sul, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Fernanda E Gandolfi
- Newborn Section, Child Health and Adolescent Medicine Graduate Program, Department of Pediatrics, Universidade Federal do Rio Grande do Sul, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Carolina Panceri
- Newborn Section, Child Health and Adolescent Medicine Graduate Program, Department of Pediatrics, Universidade Federal do Rio Grande do Sul, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Renato S Procianoy
- Newborn Section, Child Health and Adolescent Medicine Graduate Program, Department of Pediatrics, Universidade Federal do Rio Grande do Sul, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| |
Collapse
|
4
|
Morin C, Faure F, Mollet J, Guenoun D, Heydari-Olya A, Sautet I, Diao S, Faivre V, Pansiot J, Tabet L, Hua J, Schwendimann L, Mokhtari A, Martin-Rosique R, Chadi S, Laforge M, Demené C, Delahaye-Duriez A, Diaz-Heijtz R, Fleiss B, Matrot B, Auger S, Tanter M, Van Steenwinckel J, Gressens P, Bokobza C. C-section and systemic inflammation synergize to disrupt the neonatal gut microbiota and brain development in a model of prematurity. Brain Behav Immun 2025; 123:824-837. [PMID: 39442636 DOI: 10.1016/j.bbi.2024.10.023] [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: 10/31/2023] [Revised: 10/07/2024] [Accepted: 10/20/2024] [Indexed: 10/25/2024] Open
Abstract
Infants born very preterm (below 28 weeks of gestation) are at high risk of developing neurodevelopmental disorders, such as intellectual deficiency, autism spectrum disorders, and attention deficit. Preterm birth often occurs in the context of perinatal systemic inflammation due to chorioamnionitis and postnatal sepsis. In addition, C-section is often performed for very preterm neonates to avoid hypoxia during a vaginal delivery. We have developed and characterized a mouse model based on intraperitoneal injections of IL-1β between postnatal days one and five to reproduce perinatal systemic inflammation. This model replicates several neuropathological, brain imaging, and behavioral deficits observed in preterm infants. We hypothesized that C-sections could synergize with systemic inflammation to induce more severe brain abnormalities. We observed that C-sections significantly exacerbated the deleterious effects of IL-1β on reduced gut microbial diversity, increased levels of circulating peptidoglycans, abnormal microglia/macrophage reactivity, impaired myelination, and reduced functional connectivity in the brain relative to vaginal delivery plus intraperitoneal saline. These data demonstrate the deleterious synergistic effects of C-section and neonatal systemic inflammation on brain maldevelopment and malfunction, two conditions frequently observed in very preterm infants, who are at high risk of developing neurodevelopmental disorders.
Collapse
Affiliation(s)
- Cécile Morin
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France; Department of Obstetrics and Gynecology, AP-HP, Robert Debré Hospital, 75019 Paris, France
| | - Flora Faure
- Physics for Medicine Paris, Inserm, ESPCI Paris-PSL, CNRS, 75015 Paris, France
| | - Julie Mollet
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France
| | - David Guenoun
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France; Department of Pharmacy, AP-HP, Robert Debré Hospital, 75019 Paris, France
| | | | - Irvin Sautet
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France
| | - Sihao Diao
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France; Fudan University, Department of Neonatology, Children's Hospital of Fudan University, 201102 Shanghai, China
| | - Valérie Faivre
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France
| | - Julien Pansiot
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France
| | - Lara Tabet
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France
| | - Jennifer Hua
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France
| | | | - Amazigh Mokhtari
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France
| | - Rebeca Martin-Rosique
- INRAE, Université Paris-Saclay, AgroParisTech, UMR1319 Micalis Institute, 78352 Jouy-en-Josas, France
| | - Sead Chadi
- INRAE, Université Paris-Saclay, AgroParisTech, UMR1319 Micalis Institute, 78352 Jouy-en-Josas, France
| | - Mireille Laforge
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France
| | - Charlie Demené
- Physics for Medicine Paris, Inserm, ESPCI Paris-PSL, CNRS, 75015 Paris, France
| | - Andrée Delahaye-Duriez
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France; UFR Santé Médecine et Biologie Humaine, Université Sorbonne Paris Nord, 93000 Bobigny, France; Unité Fonctionnelle de Médecine Génomique et Génétique Clinique, Hôpital Jean Verdier, Hôpitaux Universitaires Paris Seine Saint-Denis, Assistance Publique des Hôpitaux de Paris, 93140 Bondy, France
| | | | - Bobbi Fleiss
- School of Health and Biomedical Sciences, STEM College, RMIT University, Bundoora, Melbourne, Victoria 3083, Australia
| | - Boris Matrot
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France
| | - Sandrine Auger
- INRAE, Université Paris-Saclay, AgroParisTech, UMR1319 Micalis Institute, 78352 Jouy-en-Josas, France
| | - Mickael Tanter
- Physics for Medicine Paris, Inserm, ESPCI Paris-PSL, CNRS, 75015 Paris, France
| | | | - Pierre Gressens
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France
| | - Cindy Bokobza
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France.
| |
Collapse
|
5
|
Hou L, Zhao J, Yin L, Dai L, Deng H, Jiang L. Brain injury in premature infants may be related to abnormal colonization of early gut microbiome. BMC Microbiol 2024; 24:483. [PMID: 39558267 PMCID: PMC11575211 DOI: 10.1186/s12866-024-03643-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 11/07/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Premature infants are more prone to brain injuries owing to incomplete nervous system development and poor adaptation outside the mother's body. Without timely intervention, premature infants with brain injuries often develop intellectual disabilities, causing significant burdens on families and the society. Multiple studies have shown that gut dysbiosis can affect the nervous system, and vice versa. This study aimed to explore the changes in gut microbiota of typical premature infants and those with brain injuries on the third and seventh days after birth using 16 S rRNA technology. METHODS Fecal samples from typical premature infants (non-brain injury group, n = 17) and those with brain injuries (brain injury group, n = 21) were collected on days 1, 3, and 7 after birth for 16 S rRNA sequencing. Alpha diversity analysis was used to evaluate the diversity of gut microbiome. LEfSe and DESeq2 were used to analyze of the microorganisms' characteristics and differentiate the microorganisms between the two groups. RESULTS At the phylum level, Firmicutes, Proteobacteria, and Actinobacteria were the dominant flora in both groups. At the genus level, the proportion of Enterococcus in fecal samples of the brain injury group was higher than that of the non-brain injury group on day three after birth; however, the opposite was observed on day seven. Rothia and Lactobacillales were characteristic bacteria of the non-brain injury group on days three and seven after birth, whereas Enterococcus and Bifidobacteria were characteristic bacteria of the brain injury group on days three and seven after birth, respectively. Three days after birth, the Shannon and Simpson indices of the non-brain injury group were significantly higher than those of the brain injury group. CONCLUSION Premature infants with brain injuries have a unique gut microbiota that is different from that of typical premature infants, indicating correlation between brain injuries and gut microbiota.
Collapse
Affiliation(s)
- Li Hou
- Department of Neonatology, Affiliated Hospital of North Sichuan Medical College, No.1, Mao Yuan South Road, Shunqing District, Nanchong, 637000, Sichuan, China
| | - Jing Zhao
- Department of Neonatology, Affiliated Hospital of North Sichuan Medical College, No.1, Mao Yuan South Road, Shunqing District, Nanchong, 637000, Sichuan, China.
| | - Linlin Yin
- Department of Neonatology, Affiliated Hospital of North Sichuan Medical College, No.1, Mao Yuan South Road, Shunqing District, Nanchong, 637000, Sichuan, China
| | - Lu Dai
- Department of Neonatology, Affiliated Hospital of North Sichuan Medical College, No.1, Mao Yuan South Road, Shunqing District, Nanchong, 637000, Sichuan, China
| | - Hong Deng
- Department of Neonatology, Affiliated Hospital of North Sichuan Medical College, No.1, Mao Yuan South Road, Shunqing District, Nanchong, 637000, Sichuan, China
| | - Lin Jiang
- Department of Neonatology, Affiliated Hospital of North Sichuan Medical College, No.1, Mao Yuan South Road, Shunqing District, Nanchong, 637000, Sichuan, China
| |
Collapse
|
6
|
Kim YH, Lee TY, Kim HY, Jeong SJ, Han JH, Shin JE, Lee JH, Kang CM. Natal factors influencing newborn's oral microbiome diversity. Sci Rep 2024; 14:28161. [PMID: 39548168 PMCID: PMC11568190 DOI: 10.1038/s41598-024-78609-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 11/02/2024] [Indexed: 11/17/2024] Open
Abstract
The early microbiota of neonates is crucial for developing the postnatal immune system and establishing normal physiological, metabolic, and neurological functions. This study aimed to investigate the factors influencing the diversity of the neonatal oral microbiome, including mother-to-newborn microbial transmission. The study includes a prospective cohort comprising 73 mothers and 87 neonates and a retrospective cohort comprising 991 mothers and 1,121 neonates. Samples from the maternal cervix and neonatal gastric, bronchial, and oral cavities were analyzed using culture-based methods. Neonatal oral swab samples were also analyzed using 16S rRNA gene sequencing to characterize microbial diversity and composition. Similar genera were detected in the neonatal gastric, bronchial, and oral samples, and the neonatal gastric culture was the most similar to the maternal cervical culture. In addition to mother-to-newborn microbial transmission, various natal factors including birth type, birth weight, delivery mode, maternal chorioamnionitis, maternal diabetes and the presence of microbes in other sites influenced neonatal oral microbiome diversity. Among these factors, the birth type was the most significant, and preterm neonates exhibited decreased oral microbiome diversity, with fewer beneficial bacteria and more pathogens. These findings could serve as a baseline for research on the establishment of the oral microbiota in preterm neonates and its health implications.
Collapse
Affiliation(s)
- Yoon-Hee Kim
- Department of Pediatric Dentistry, Yonsei University College of Dentistry, 50-1 Yonsei-ro, Seoul, 03722, Republic of Korea
| | - Tae Yang Lee
- Department of Applied Life Science, The Graduate School, Yonsei University, Seoul, Republic of Korea
- Oral Science Research Center, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | | | - Su Jin Jeong
- Statistics Support Part, Medical Science Research Institute, Kyung Hee University Medical Center, Seoul, Republic of Korea
| | - Jung Ho Han
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jeong Eun Shin
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae-Ho Lee
- Department of Pediatric Dentistry, Yonsei University College of Dentistry, 50-1 Yonsei-ro, Seoul, 03722, Republic of Korea
- Oral Science Research Center, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - Chung-Min Kang
- Department of Pediatric Dentistry, Yonsei University College of Dentistry, 50-1 Yonsei-ro, Seoul, 03722, Republic of Korea.
- Oral Science Research Center, Yonsei University College of Dentistry, Seoul, Republic of Korea.
| |
Collapse
|
7
|
Magnusson A, Jabbari Shiadeh SM, Ardalan M, Swolin-Eide D, Elfvin A. Gut microbiota differences in five-year-old children that were born preterm with a history of necrotizing enterocolitis: A pilot trial. iScience 2024; 27:110325. [PMID: 39055941 PMCID: PMC11269947 DOI: 10.1016/j.isci.2024.110325] [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: 12/18/2023] [Revised: 04/22/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024] Open
Abstract
The study explores the long-term effects of necrotizing enterocolitis (NEC) on gut microbiota in preterm infants by analyzing stool samples from 5-year-old children using shotgun metagenomic sequencing. It compares children with a history of NEC, treated surgically or medically, to preterm controls without NEC. Findings reveal persistent gut microbiota dysbiosis in NEC children, with reduced species diversity and evenness, especially in those treated surgically. The surgical NEC group had a lower Shannon index, indicating less microbial diversity. Significant differences in taxonomic profiles were observed, mainly influenced by surgical treatment. These results underscore the lasting impact of NEC and its treatment on gut microbiota, suggesting a need for strategies addressing long-term dysbiosis.
Collapse
Affiliation(s)
- Amanda Magnusson
- Department of Pediatrics, Institution of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Department of Pediatrics, The Queen Silvia Children’s Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Seyedeh Marziyeh Jabbari Shiadeh
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Medicine, Translational Neuropsychiatry Unit, Aarhus University, Aarhus, Denmark
| | - Maryam Ardalan
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Medicine, Translational Neuropsychiatry Unit, Aarhus University, Aarhus, Denmark
| | - Diana Swolin-Eide
- Department of Pediatrics, Institution of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Department of Pediatrics, The Queen Silvia Children’s Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anders Elfvin
- Department of Pediatrics, Institution of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Department of Pediatrics, The Queen Silvia Children’s Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
8
|
Radocchia G, Brunetti F, Marazzato M, Totino V, Neroni B, Bonfiglio G, Conte AL, Pantanella F, Ciolli P, Schippa S. Women Skin Microbiota Modifications during Pregnancy. Microorganisms 2024; 12:808. [PMID: 38674752 PMCID: PMC11051999 DOI: 10.3390/microorganisms12040808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 03/27/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
Several studies have shown fluctuations in the maternal microbiota at various body sites (gut, oral cavity, and vagina). The skin microbiota plays an important role in our health, but studies on the changes during pregnancy are limited. Quantitative and qualitative variations in the skin microbiota in pregnant woman could indeed play important roles in modifying the immune and inflammatory responses of the host. These alterations could induce inflammatory disorders affecting the individual's dermal properties, and could potentially predict infant skin disorder in the unborn. The present study aimed to characterize skin microbiota modifications during pregnancy. For this purpose, skin samples were collected from 52 pregnant women in the first, second, and third trimester of non-complicated pregnancies and from 17 age- and sex-matched healthy controls. The skin microbiota composition was assessed by next generation sequencing (NGS) of the V3-V4 region of the bacterial rRNA 16S. Our results indicate that from the first to the third trimester of pregnancy, changes occur in the composition of the skin microbiota, microbial interactions, and various metabolic pathways. These changes could play a role in creating more advantageous conditions for fetal growth.
Collapse
Affiliation(s)
- Giulia Radocchia
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy; (F.B.); (M.M.); (V.T.); (B.N.); (G.B.); (A.L.C.); (F.P.); (S.S.)
| | - Francesca Brunetti
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy; (F.B.); (M.M.); (V.T.); (B.N.); (G.B.); (A.L.C.); (F.P.); (S.S.)
| | - Massimiliano Marazzato
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy; (F.B.); (M.M.); (V.T.); (B.N.); (G.B.); (A.L.C.); (F.P.); (S.S.)
| | - Valentina Totino
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy; (F.B.); (M.M.); (V.T.); (B.N.); (G.B.); (A.L.C.); (F.P.); (S.S.)
- Policlinico Luigi Di Liegro, 00148 Rome, Italy
| | - Bruna Neroni
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy; (F.B.); (M.M.); (V.T.); (B.N.); (G.B.); (A.L.C.); (F.P.); (S.S.)
- Diagnostic Medicine and Radiology, UOC Clinical Pathology, Policlinico Umberto I Hospital, 00161 Rome, Italy
| | - Giulia Bonfiglio
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy; (F.B.); (M.M.); (V.T.); (B.N.); (G.B.); (A.L.C.); (F.P.); (S.S.)
- Diagnostic Medicine and Radiology, UOC Clinical Pathology, Policlinico Umberto I Hospital, 00161 Rome, Italy
| | - Antonietta Lucia Conte
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy; (F.B.); (M.M.); (V.T.); (B.N.); (G.B.); (A.L.C.); (F.P.); (S.S.)
| | - Fabrizio Pantanella
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy; (F.B.); (M.M.); (V.T.); (B.N.); (G.B.); (A.L.C.); (F.P.); (S.S.)
| | - Paola Ciolli
- Department of Maternal Infantile and Urological Sciences, Policlinico Umberto I Hospital, Sapienza University of Rome, 00185 Rome, Italy;
| | - Serena Schippa
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy; (F.B.); (M.M.); (V.T.); (B.N.); (G.B.); (A.L.C.); (F.P.); (S.S.)
| |
Collapse
|
9
|
Bhatia Z, Kumar S, Seshadri S. Composition and interaction of maternal microbiota with immune mediators during pregnancy and their outcome: A narrative review. Life Sci 2024; 340:122440. [PMID: 38278350 DOI: 10.1016/j.lfs.2024.122440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/26/2023] [Accepted: 01/14/2024] [Indexed: 01/28/2024]
Abstract
The connection between maternal microbiota and infant health has been greatly garnered interest for therapeutic purposes. The early resident microbiota perpetually exhibits much more flexibility as compared to that of the adults, and therefore, constant need of understanding the infant as well as maternal microbiota and their implications however has increased. In this review, we focus mainly on the diversity of overall maternal microbiota including the gut, vaginal, colostrum microbiota and how inflammatory markers fluctuate throughout the normal pregnancy as well in pregnancy with complications. The maternal body undergoes a cascade of physiological changes including hormonal, immunological and metabolic events to support the fetal development. These changes at the time of pregnancy have been correlated with alteration in the composition and diversity of maternal microbiota. Along with alteration in microbiome, the levels of circulatory cytokines fluctuate by complex network of inflammation, in order to prevent the fetal allograft throughout the pregnancy. The dynamic relationship of gut microbiota with the host and its immune system allows one to have greater insights of their role in pregnancy and newborn's health. Emerging evidence suggests that the vertical transmission of bacterial community from mother to newborn may begin in-utero which contributes in developing the immune system and infant gut microbiota.
Collapse
Affiliation(s)
- Zeel Bhatia
- Institute of Science, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Sunny Kumar
- Institute of Science, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Sriram Seshadri
- Institute of Science, Nirma University, Ahmedabad, Gujarat 382481, India.
| |
Collapse
|
10
|
VanOrmer M, Thompson M, Thoene M, Riethoven JJ, Natarajan SK, Hanson C, Anderson-Berry A. The impact of iron supplementation on the preterm neonatal gut microbiome: A pilot study. PLoS One 2024; 19:e0297558. [PMID: 38381745 PMCID: PMC10880995 DOI: 10.1371/journal.pone.0297558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 01/08/2024] [Indexed: 02/23/2024] Open
Abstract
OBJECTIVE The gastrointestinal microbiome in preterm infants exhibits significant influence on optimal outcomes-with dysbiosis shown to substantially increase the risk of the life-threatening necrotizing enterocolitis. Iron is a vital nutrient especially during the perinatal window of rapid hemoglobin production, tissue growth, and foundational neurodevelopment. However, excess colonic iron exhibits potent oxidation capacity and alters the gut microbiome-potentially facilitating the proliferation of pathological bacterial strains. Breastfed preterm infants routinely receive iron supplementation starting 14 days after delivery and are highly vulnerable to morbidities associated with gastrointestinal dysbiosis. Therefore, we set out to determine if routine iron supplementation alters the preterm gut microbiome. METHODS After IRB approval, we collected stool specimens from 14 infants born <34 weeks gestation in the first, second, and fourth week of life to assess gut microbiome composition via 16S rRNA sequencing. RESULTS We observed no significant differences in either phyla or key genera relative abundance between pre- and post-iron timepoints. We observed notable shifts in infant microbiome composition based on season of delivery. CONCLUSION Though no obvious indication of iron-induced dysbiosis was observed in this unique study in the setting of prematurity, further investigation in a larger sample is warranted to fully understand iron's impact on the gastrointestinal milieu.
Collapse
Affiliation(s)
- Matthew VanOrmer
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Maranda Thompson
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Melissa Thoene
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Jean-Jack Riethoven
- Nebraska Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Sathish Kumar Natarajan
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Corrine Hanson
- College of Allied Health Professions, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Ann Anderson-Berry
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, United States of America
| |
Collapse
|
11
|
Samarra A, Flores E, Bernabeu M, Cabrera-Rubio R, Bäuerl C, Selma-Royo M, Collado MC. Shaping Microbiota During the First 1000 Days of Life. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1449:1-28. [PMID: 39060728 DOI: 10.1007/978-3-031-58572-2_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
Given that the host-microbe interaction is shaped by the immune system response, it is important to understand the key immune system-microbiota relationship during the period from conception to the first years of life. The present work summarizes the available evidence concerning human reproductive microbiota, and also, the microbial colonization during early life, focusing on the potential impact on infant development and health outcomes. Furthermore, we conclude that some dietary strategies including specific probiotics and other-biotics could become potentially valuable tools to modulate the maternal-neonatal microbiota during this early critical window of opportunity for targeted health outcomes throughout the entire lifespan.
Collapse
Affiliation(s)
- Anna Samarra
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Paterna-Valencia, Spain
| | - Eduard Flores
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Paterna-Valencia, Spain
| | - Manuel Bernabeu
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Paterna-Valencia, Spain
| | - Raul Cabrera-Rubio
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Paterna-Valencia, Spain
| | - Christine Bäuerl
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Paterna-Valencia, Spain
| | - Marta Selma-Royo
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Paterna-Valencia, Spain
| | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Paterna-Valencia, Spain.
| |
Collapse
|
12
|
Koren O, Konnikova L, Brodin P, Mysorekar IU, Collado MC. The maternal gut microbiome in pregnancy: implications for the developing immune system. Nat Rev Gastroenterol Hepatol 2024; 21:35-45. [PMID: 38097774 DOI: 10.1038/s41575-023-00864-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/20/2023] [Indexed: 01/04/2024]
Abstract
The gut microbiome has important roles in host metabolism and immunity, and microbial dysbiosis affects human physiology and health. Maternal immunity and microbial metabolites during pregnancy, microbial transfer during birth, and transfer of immune factors, microorganisms and metabolites via breastfeeding provide critical sources of early-life microbial and immune training, with important consequences for human health. Only a few studies have directly examined the interactions between the gut microbiome and the immune system during pregnancy, and the subsequent effect on offspring development. In this Review, we aim to describe how the maternal microbiome shapes overall pregnancy-associated maternal, fetal and early neonatal immune systems, focusing on the existing evidence and highlighting current gaps to promote further research.
Collapse
Affiliation(s)
- Omry Koren
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Liza Konnikova
- Department of Paediatrics and Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Petter Brodin
- Department of Immunology and Inflammation, Imperial College London, London, UK
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Indira U Mysorekar
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Valencia, Spain.
| |
Collapse
|
13
|
Jain A, Meshram RJ, Lohiya S, Patel A, Kaplish D. Exploring the Microbial Landscape of Neonatal Skin Flora: A Comprehensive Review. Cureus 2024; 16:e52972. [PMID: 38406113 PMCID: PMC10894447 DOI: 10.7759/cureus.52972] [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: 01/12/2024] [Accepted: 01/26/2024] [Indexed: 02/27/2024] Open
Abstract
This comprehensive review explores the intricate landscape of the neonatal skin microbiome, shedding light on its dynamic composition, developmental nuances, and influential factors. The neonatal period represents a critical window during which microbial colonization significantly impacts local skin health and the foundational development of the immune system. Factors such as mode of delivery and gestational age underscore the vulnerability of neonates to disruptions in microbial establishment. Key findings emphasize the broader systemic implications of the neonatal skin microbiome, extending beyond immediate health outcomes to influence susceptibility to infections, allergies, and immune-related disorders. This review advocates for a paradigm shift in neonatal care, proposing strategies to preserve and promote a healthy skin microbiome for long-term health benefits. The implications of this research extend to public health, where interventions targeting the neonatal skin microbiome could potentially mitigate diseases originating in early life. As we navigate the intersection of research and practical applications, bridging the gap between knowledge and implementation becomes imperative for translating these findings into evidence-based practices and improving neonatal well-being on a broader scale.
Collapse
Affiliation(s)
- Aditya Jain
- Pediatrics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Revat J Meshram
- Pediatrics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Sham Lohiya
- Pediatrics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Ankita Patel
- Pediatrics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Divyanshi Kaplish
- Pediatrics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
14
|
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
|
15
|
Vanzele PAR, Sparvoli LG, de Camargo PP, Tragante CR, Beozzo GPNS, Krebs VLJ, Cortez RV, Taddei CR. Establishment of oral microbiome in very low birth weight infants during the first weeks of life and the impact of oral diet implementation. PLoS One 2023; 18:e0295962. [PMID: 38100452 PMCID: PMC10723731 DOI: 10.1371/journal.pone.0295962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 12/02/2023] [Indexed: 12/17/2023] Open
Abstract
Very low birth weight (VLBW) infants, mostly preterm, have many barriers to feeding directly from the mother's breast, and need to be fed alternatively. Feeding is a major influencer in oral microbial colonization, and this colonization in early life is crucial for the promotion of human health. Therefore, this research aimed to observe the establishment of oral microbiome in VLBW infants during their first month of life through hospitalization, and to verify the impact caused by the implementation of oral diet on the colonization of these newborns. We included 23 newborns followed during hospitalization and analyzed saliva samples collected weekly, using 16S rRNA gene sequencing. We observed a significant decrease in richness and diversity and an increase in dominance over time (q-value < 0.05). The oral microbiome is highly dynamic during the first weeks of life, and beta diversity suggests a microbial succession in early life. The introduction of oral diet does not change the community structure, but affects the abundance, especially of Streptococcus. Our results indicate that although time is related to significant changes in the oral microbial profile, oral feeding benefits genera that will remain colonizers throughout the host's life.
Collapse
Affiliation(s)
- Pedro A. R. Vanzele
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Luiz Gustavo Sparvoli
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Patricia P. de Camargo
- Neonatal Intensive Care Center, Children’s Institute, Hospital das Clínicas, São Paulo Medical School, University of São Paulo, São Paulo, SP, Brazil
| | - Carla R. Tragante
- Neonatal Intensive Care Center, Children’s Institute, Hospital das Clínicas, São Paulo Medical School, University of São Paulo, São Paulo, SP, Brazil
| | - Glenda P. N. S. Beozzo
- Neonatal Intensive Care Center, Children’s Institute, Hospital das Clínicas, São Paulo Medical School, University of São Paulo, São Paulo, SP, Brazil
| | - Vera L. J. Krebs
- Neonatal Intensive Care Center, Children’s Institute, Hospital das Clínicas, São Paulo Medical School, University of São Paulo, São Paulo, SP, Brazil
| | - Ramon V. Cortez
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Carla R. Taddei
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
- School of Arts, Sciences and Humanity, University of São Paulo, São Paulo, SP, Brazil
- Division of Clinical Laboratory, University Hospital ‐ University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
16
|
Xiu W, Lin J, Hu Y, Tang H, Wu S, Yang C. Assessing multiple factors affecting the gut microbiome structure of very preterm infants. Braz J Med Biol Res 2023; 56:e13186. [PMID: 38088676 PMCID: PMC10712283 DOI: 10.1590/1414-431x2023e13186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 10/30/2023] [Indexed: 12/18/2023] Open
Abstract
The composition and diversity of the gut microbiota are essential for the health and development of the immune system of infants. However, there is limited information on factors that influence the gut microbiota of very preterm infants. In this study, we analyzed factors that affect the gut microbiota of very preterm infants. The stool samples from 64 very preterm infants with a gestational age less than 32 weeks were collected for 16S rRNA gene sequencing. The infants were divided according to the delivery mode, antibiotic use during pregnancy, and feeding methods. The abundance of Proteobacteria was high in both cesarean (92.7%) and spontaneous (55.5%) delivery groups and then shifted to Firmicutes after the first week of birth. In addition, Proteobacteria was also the dominant phylum of infant gut microbiome for mothers with antibiotic use, with more than 50% after the first week of birth. In comparison, the dominant phylum for mothers without antibiotic use was Firmicutes. Proteobacteria level was also high in breastfeeding and mixed-feeding groups, consisting of more than 90% of the community. By contrast, Proteobacteria was the dominant phylum at the first week of birth but then shifted to Firmicutes for the formula-fed group. The alterations of gut microbiota in infants can affect their health condition during growth. This study confirmed that the different feeding types, delivery modes, and use of antibiotics during pregnancy can significantly affect the composition of the gut microbiota of very preterm infants.
Collapse
Affiliation(s)
- Wenlong Xiu
- Department of Neonatology, Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Jiajia Lin
- Department of Neonatology, Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Yanhua Hu
- Department of Neonatology, Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Heng Tang
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Shuangchan Wu
- Institute of Medical Research, Northwestern Polytechnical University, Xian, Shanxi Province, China
| | - Changyi Yang
- Department of Neonatology, Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian Province, China
| |
Collapse
|
17
|
Farias-Jofre M, Romero R, Galaz J, Xu Y, Miller D, Garcia-Flores V, Arenas-Hernandez M, Winters AD, Berkowitz BA, Podolsky RH, Shen Y, Kanninen T, Panaitescu B, Glazier CR, Pique-Regi R, Theis KR, Gomez-Lopez N. Blockade of IL-6R prevents preterm birth and adverse neonatal outcomes. EBioMedicine 2023; 98:104865. [PMID: 37944273 PMCID: PMC10665693 DOI: 10.1016/j.ebiom.2023.104865] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/11/2023] [Accepted: 10/19/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Preterm birth preceded by spontaneous preterm labour often occurs in the clinical setting of sterile intra-amniotic inflammation (SIAI), a condition that currently lacks treatment. METHODS Proteomic and scRNA-seq human data were analysed to evaluate the role of IL-6 and IL-1α in SIAI. A C57BL/6 murine model of SIAI-induced preterm birth was developed by the ultrasound-guided intra-amniotic injection of IL-1α. The blockade of IL-6R by using an aIL-6R was tested as prenatal treatment for preterm birth and adverse neonatal outcomes. QUEST-MRI evaluated brain oxidative stress in utero. Targeted transcriptomic profiling assessed maternal, foetal, and neonatal inflammation. Neonatal biometrics and neurodevelopment were tested. The neonatal gut immune-microbiome was evaluated using metagenomic sequencing and immunophenotyping. FINDINGS IL-6 plays a critical role in the human intra-amniotic inflammatory response, which is associated with elevated concentrations of the alarmin IL-1α. Intra-amniotic injection of IL-1α resembles SIAI, inducing preterm birth (7% vs. 50%, p = 0.03, Fisher's exact test) and neonatal mortality (18% vs. 56%, p = 0.02, Mann-Whitney U-test). QUEST-MRI revealed no foetal brain oxidative stress upon in utero IL-1α exposure (p > 0.05, mixed linear model). Prenatal treatment with aIL-6R abrogated IL-1α-induced preterm birth (50% vs. 7%, p = 0.03, Fisher's exact test) by dampening inflammatory processes associated with the common pathway of labour. Importantly, aIL-6R reduces neonatal mortality (56% vs. 22%, p = 0.03, Mann-Whitney U-test) by crossing from the mother to the amniotic cavity, dampening foetal organ inflammation and improving growth. Beneficial effects of prenatal IL-6R blockade carried over to neonatal life, improving survival, growth, neurodevelopment, and gut immune homeostasis. INTERPRETATION IL-6R blockade can serve as a strategy to treat SIAI, preventing preterm birth and adverse neonatal outcomes. FUNDING NICHD/NIH/DHHS, Contract HHSN275201300006C. WSU Perinatal Initiative in Maternal, Perinatal and Child Health.
Collapse
Affiliation(s)
- Marcelo Farias-Jofre
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA; Division of Obstetrics and Gynecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Roberto Romero
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, USA; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA.
| | - Jose Galaz
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA; Division of Obstetrics and Gynecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Yi Xu
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Derek Miller
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA; Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, USA
| | - Valeria Garcia-Flores
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA; Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, USA
| | - Marcia Arenas-Hernandez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Andrew D Winters
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, USA; Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MO, USA
| | - Bruce A Berkowitz
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine; Detroit, MI, USA
| | - Robert H Podolsky
- Division of Biostatistics and Design Methodology, Center for Translational Research, Children's National Hospital, Silver Spring, MD, USA
| | - Yimin Shen
- Department of Radiology, School of Medicine, Wayne State University School of Medicine, Detroit, MI, USA
| | - Tomi Kanninen
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Bogdan Panaitescu
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Catherine R Glazier
- UCD School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Roger Pique-Regi
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA; Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Kevin R Theis
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA; Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MO, USA
| | - Nardhy Gomez-Lopez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA; Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, USA; Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MO, USA; Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
18
|
Luoto R, Pärtty A, Vogt JK, Rautava S, Isolauri E. Reversible aberrancies in gut microbiome of moderate and late preterm infants: results from a randomized, controlled trial. Gut Microbes 2023; 15:2283913. [PMID: 38010080 PMCID: PMC10730193 DOI: 10.1080/19490976.2023.2283913] [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: 06/15/2023] [Accepted: 11/12/2023] [Indexed: 11/29/2023] Open
Abstract
The aim of this study was to obtain insight into the composition and function of the deviant gut microbiome throughout infancy in children born moderately and late preterm and their response to microbiome modulation. We characterized the longitudinal development of the gut microbiome from birth to the age of 12 months by metagenomic sequencing in 43 moderate and late preterm children participating in a randomized, controlled trial (ClinicalTrials.gov/no.NCT00167700) assessing the impact of a probiotic (Lactobacillus rhamnosus GG, ATCC 53,103, currently Lacticaseibacillus rhamnosus GG) and a prebiotic (galacto-oligosaccharide and polydextrose mixture, 1:1) intervention as compared to a placebo administered from 3 to 60 days of life. In addition, 9 full-term, vaginally delivered, breast-fed infants, who remained healthy long-term were included as references. Significant differences in taxonomy, but not in functional potential, were found when comparing the gut microbiome composition of preterm and full-term infants during the first month of life. However, the gut microbiome of preterm infants resembled that of full-term infants by 6 months age. Probiotic and prebiotic treatments were found to mitigate the shift in the microbiome of preterm infants by accelerating Bifidobacteria-dominated gut microbiome in beta diversity analysis. This study provides intriguing information regarding the establishment of the gut microbiome in children born moderately and late preterm, representing the majority of children born preterm. Specific pro- and prebiotics may reverse the proinflammatory gut microbiome composition during the vulnerable period, when the microbiome is low in resilience and susceptible to environmental exposure and simultaneously promotes immunological and metabolic maturation.
Collapse
Affiliation(s)
- Raakel Luoto
- Department of Pediatrics and Adolescent medicine, Turku University Hospital, Turku, Finland
- Institute of Clinical Medicine, University of Turku, Turku, Finland
| | - Anna Pärtty
- Department of Pediatrics and Adolescent medicine, Turku University Hospital, Turku, Finland
- Institute of Clinical Medicine, University of Turku, Turku, Finland
| | | | - Samuli Rautava
- Institute of Clinical Medicine, University of Turku, Turku, Finland
- New Children’s Hospital, Helsinki University Hospital, Helsinki, Finland
| | - Erika Isolauri
- Department of Pediatrics and Adolescent medicine, Turku University Hospital, Turku, Finland
- Institute of Clinical Medicine, University of Turku, Turku, Finland
| |
Collapse
|
19
|
Barreiros-Mota I, R. Araújo J, Marques C, Sousa L, Morais J, Castela I, Faria A, Neto MT, Cordeiro-Ferreira G, Virella D, Pita A, Pereira-da-Silva L, Calhau C. Changes in Microbiota Profile in the Proximal Remnant Intestine in Infants Undergoing Surgery Requiring Enterostomy. Microorganisms 2023; 11:2482. [PMID: 37894140 PMCID: PMC10609405 DOI: 10.3390/microorganisms11102482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 09/29/2023] [Accepted: 09/30/2023] [Indexed: 10/29/2023] Open
Abstract
Early-life gut dysbiosis has been associated with an increased risk of inflammatory, metabolic, and immune diseases later in life. Data on gut microbiota changes in infants undergoing intestinal surgery requiring enterostomy are scarce. This prospective cohort study examined the enterostomy effluent of 29 infants who underwent intestinal surgery due to congenital malformations of the gastrointestinal tract, necrotizing enterocolitis, or spontaneous intestinal perforation. Initial effluent samples were collected immediately after surgery and final effluent samples were collected three weeks later. Gut microbiota composition was analysed using real-time PCR and 16S rRNA gene sequencing. Three weeks after surgery, an increase in total bacteria number (+21%, p = 0.026), a decrease in Staphylococcus (-21%, p = 0.002) and Candida spp. (-16%, p = 0.045), and an increase in Lactobacillus (+3%, p = 0.045) and in less abundant genera belonging to the Enterobacteriales family were found. An increase in alpha diversity (Shannon's and Simpson's indexes) and significant alterations in beta diversity were observed. A correlation of necrotizing enterocolitis with higher Staphylococcus abundance and higher alpha diversity was also observed. H2-blockers and/or proton pump inhibitor therapy were positively correlated with a higher total bacteria number. In conclusion, these results suggest that positive changes occur in the gut microbiota profile of infants three weeks after intestinal surgery.
Collapse
Affiliation(s)
- Inês Barreiros-Mota
- Nutrition & Metabolism Department, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal; (I.B.-M.); (J.R.A.); (C.M.); (L.S.); (I.C.); (A.F.)
- CHRC—Comprehensive Health Research Centre, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal; (J.M.); (M.T.N.); (L.P.-d.-S.)
| | - João R. Araújo
- Nutrition & Metabolism Department, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal; (I.B.-M.); (J.R.A.); (C.M.); (L.S.); (I.C.); (A.F.)
- Nutrition & Metabolism Department, CINTESIS@RISE, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal
| | - Cláudia Marques
- Nutrition & Metabolism Department, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal; (I.B.-M.); (J.R.A.); (C.M.); (L.S.); (I.C.); (A.F.)
- Nutrition & Metabolism Department, CINTESIS@RISE, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal
| | - Laura Sousa
- Nutrition & Metabolism Department, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal; (I.B.-M.); (J.R.A.); (C.M.); (L.S.); (I.C.); (A.F.)
| | - Juliana Morais
- CHRC—Comprehensive Health Research Centre, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal; (J.M.); (M.T.N.); (L.P.-d.-S.)
| | - Inês Castela
- Nutrition & Metabolism Department, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal; (I.B.-M.); (J.R.A.); (C.M.); (L.S.); (I.C.); (A.F.)
- CHRC—Comprehensive Health Research Centre, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal; (J.M.); (M.T.N.); (L.P.-d.-S.)
| | - Ana Faria
- Nutrition & Metabolism Department, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal; (I.B.-M.); (J.R.A.); (C.M.); (L.S.); (I.C.); (A.F.)
- CHRC—Comprehensive Health Research Centre, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal; (J.M.); (M.T.N.); (L.P.-d.-S.)
- Nutrition & Metabolism Department, CINTESIS@RISE, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal
| | - Maria Teresa Neto
- CHRC—Comprehensive Health Research Centre, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal; (J.M.); (M.T.N.); (L.P.-d.-S.)
- Neonatal Intensive Care Unit, Department of Pediatrics, Hospital Dona Estefânia, Centro Hospitalar Universitário de Lisboa Central, 1169-045 Lisbon, Portugal; (G.C.-F.); (D.V.); (A.P.)
- Medicine of Woman, Childhood and Adolescence Academic Area, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), 1169-056 Lisbon, Portugal
| | - Gonçalo Cordeiro-Ferreira
- Neonatal Intensive Care Unit, Department of Pediatrics, Hospital Dona Estefânia, Centro Hospitalar Universitário de Lisboa Central, 1169-045 Lisbon, Portugal; (G.C.-F.); (D.V.); (A.P.)
| | - Daniel Virella
- Neonatal Intensive Care Unit, Department of Pediatrics, Hospital Dona Estefânia, Centro Hospitalar Universitário de Lisboa Central, 1169-045 Lisbon, Portugal; (G.C.-F.); (D.V.); (A.P.)
| | - Ana Pita
- Neonatal Intensive Care Unit, Department of Pediatrics, Hospital Dona Estefânia, Centro Hospitalar Universitário de Lisboa Central, 1169-045 Lisbon, Portugal; (G.C.-F.); (D.V.); (A.P.)
| | - Luís Pereira-da-Silva
- CHRC—Comprehensive Health Research Centre, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal; (J.M.); (M.T.N.); (L.P.-d.-S.)
- Neonatal Intensive Care Unit, Department of Pediatrics, Hospital Dona Estefânia, Centro Hospitalar Universitário de Lisboa Central, 1169-045 Lisbon, Portugal; (G.C.-F.); (D.V.); (A.P.)
- Medicine of Woman, Childhood and Adolescence Academic Area, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), 1169-056 Lisbon, Portugal
| | - Conceição Calhau
- Nutrition & Metabolism Department, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal; (I.B.-M.); (J.R.A.); (C.M.); (L.S.); (I.C.); (A.F.)
- Nutrition & Metabolism Department, CINTESIS@RISE, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal
| |
Collapse
|
20
|
Rahkola EN, Rautava S, Hiltunen H, Ross C, Lahti L, Isolauri E. The preterm gut microbiota and administration routes of different probiotics: a randomized controlled trial. Pediatr Res 2023; 94:1480-1487. [PMID: 37020105 PMCID: PMC10589095 DOI: 10.1038/s41390-023-02560-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 04/07/2023]
Abstract
BACKGROUND Preterm children with their aberrant gut microbiota and susceptibility to infections and inflammation constitute a considerable target group for probiotic therapy to generate the age-appropriate healthy microbiota. METHODS 68 preterm neonates were randomized into five intervention groups: Beginning from the median age of 3 days, 13 children received Lactobacillus rhamnosus GG (LGG) directly orally, and 17 via the lactating mother. 14 children received LGG with Bifidobacterium lactis Bb-12 (Bb12) orally, and 10 via the lactating mother. 14 children received placebo. The children's faecal microbiota was assessed at the age of 7 days by 16S rRNA gene sequencing. RESULTS The gut microbiota compositions of the children directly receiving the probiotic combination (LGG + Bb12) were significantly different from those of the children receiving the other intervention modes or placebo (p = 0.0012; PERMANOVA), the distinction being due to an increase in the relative abundance of Bifidobacterium animalis (P < 0.00010; ANCOM-BC), and the order Lactobacillales (P = 0.020; ANCOM-BC). CONCLUSION The connection between aberrant primary gut microbiota and a heightened risk of infectious and non-communicable diseases invites effective microbiota modulation. We show that the direct, early, and brief probiotic intervention of LGG + Bb12 109 CFU each, is sufficient to modulate the gut microbiota of the preterm neonate. IMPACT Preterm children have a higher risk of several health problems partly due to their aberrant gut microbiota. More research is needed to find a safe probiotic intervention to modify the gut microbiota of preterm children. The maternal administration route via breast milk might be safer for the newborn. In our study, the early and direct administration of the probiotic combination Lactobacillus rhamnosus GG with Bifidobacterium lactis Bb-12 increased the proportion of bifidobacteria in the preterm children's gut at the age of 7 days, but the maternal administration route was not as effective.
Collapse
Affiliation(s)
- Ella-Noora Rahkola
- Department of Clinical Sciences, Faculty of Medicine, University of Turku, Turku, Finland.
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland.
| | - Samuli Rautava
- Department of Clinical Sciences, Faculty of Medicine, University of Turku, Turku, Finland
- Department of Pediatrics, University of Helsinki and Children's Hospital, Helsinki University Hospital, Helsinki, Finland
| | - Henni Hiltunen
- Department of Clinical Sciences, Faculty of Medicine, University of Turku, Turku, Finland
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
| | - Chandler Ross
- Department of Computing, University of Turku, Turku, Finland
| | - Leo Lahti
- Department of Computing, University of Turku, Turku, Finland
| | - Erika Isolauri
- Department of Clinical Sciences, Faculty of Medicine, University of Turku, Turku, Finland
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
| |
Collapse
|
21
|
Morin C, Bokobza C, Fleiss B, Hill-Yardin EL, Van Steenwinckel J, Gressens P. Preterm Birth by Cesarean Section: The Gut-Brain Axis, a Key Regulator of Brain Development. Dev Neurosci 2023; 46:179-187. [PMID: 37717575 DOI: 10.1159/000534124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 09/11/2023] [Indexed: 09/19/2023] Open
Abstract
Understanding the long-term functional implications of gut microbial communities during the perinatal period is a bourgeoning area of research. Numerous studies have revealed the existence of a "gut-brain axis" and the impact of an alteration of gut microbiota composition in brain diseases. Recent research has highlighted how gut microbiota could affect brain development and behavior. Many factors in early life such as the mode of delivery or preterm birth could lead to disturbance in the assembly and maturation of gut microbiota. Notably, global rates of cesarean sections (C-sections) have increased in recent decades and remain important when considering premature delivery. Both preterm birth and C-sections are associated with an increased risk of neurodevelopmental disorders such as autism spectrum disorders, with neuroinflammation a major risk factor. In this review, we explore links between preterm birth by C-sections, gut microbiota alteration, and neuroinflammation. We also highlight C-sections as a risk factor for developmental disorders due to alterations in the microbiome.
Collapse
Affiliation(s)
- Cécile Morin
- Université Paris Cité, Inserm, NeuroDiderot, Paris, France
- Hôpital Robert Debré, Assistance Publique, Hôpitaux de Paris (APHP), Paris, France
| | - Cindy Bokobza
- Université Paris Cité, Inserm, NeuroDiderot, Paris, France
| | - Bobbi Fleiss
- Université Paris Cité, Inserm, NeuroDiderot, Paris, France
- School of Health and Biomedical Sciences, STEM College, RMIT University, Bundoora, Victoria, Australia
| | - Elisa L Hill-Yardin
- School of Health and Biomedical Sciences, STEM College, RMIT University, Bundoora, Victoria, Australia
| | | | - Pierre Gressens
- Université Paris Cité, Inserm, NeuroDiderot, Paris, France
- Hôpital Robert Debré, Assistance Publique, Hôpitaux de Paris (APHP), Paris, France
| |
Collapse
|
22
|
Lugli GA, Mancabelli L, Milani C, Fontana F, Tarracchini C, Alessandri G, van Sinderen D, Turroni F, Ventura M. Comprehensive insights from composition to functional microbe-based biodiversity of the infant human gut microbiota. NPJ Biofilms Microbiomes 2023; 9:25. [PMID: 37169786 PMCID: PMC10175488 DOI: 10.1038/s41522-023-00392-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 04/26/2023] [Indexed: 05/13/2023] Open
Abstract
During infancy, gut microbiota development is a crucial process involved in the establishment of microbe-host interactions which may persist throughout adulthood, and which are believed to influence host health. To fully understand the complexities of such interactions, it is essential to assess gut microbiota diversity of newborns and its associated microbial dynamics and relationships pertaining to health and disease. To explore microbial biodiversity during the first 3 years of human life, 10,935 shotgun metagenomic datasets were taxonomically and functionally classified. Microbial species distribution between infants revealed the presence of eight major Infant Community State Types (ICSTs), being dominated by 17 bacterial taxa, whose distribution was shown to correspond to the geographical origin and infant health status. In total, 2390 chromosomal sequences of the predominant taxa were reconstructed from metagenomic data and used in combination with 44,987 publicly available genomes to trace the distribution of microbial Population Subspecies (PS) within the different infant groups, revealing patterns of multistrain coexistence among ICSTs. Finally, implementation of a metagenomic- and metatranscriptomic-based metabolic profiling highlighted different enzymatic expression patterns of the gut microbiota that allowed us to acquire insights into mechanistic aspects of health-gut microbiota interplay in newborns. Comparison between metagenomic and metatranscriptomic data highlights how a complex environment like the human gut must be investigated by employing both sequencing methodologies and possibly supplemented with metabolomics approaches. While metagenomic analyses are very useful for microbial classification aimed at unveiling key players driving microbiota balances, using these data to explain functionalities of the microbiota is not always warranted.
Collapse
Affiliation(s)
- Gabriele Andrea Lugli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Leonardo Mancabelli
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Christian Milani
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Federico Fontana
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Chiara Tarracchini
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Giulia Alessandri
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Douwe van Sinderen
- APC Microbiome Institute and School of Microbiology, Bioscience Institute, National University of Ireland, T12YT20, Cork, Ireland
| | - Francesca Turroni
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy.
- Microbiome Research Hub, University of Parma, Parma, Italy.
| |
Collapse
|
23
|
Lu J, Drobyshevsky A, Lu L, Yu Y, Caplan MS, Claud EC. Microbiota from Preterm Infants Who Develop Necrotizing Enterocolitis Drives the Neurodevelopment Impairment in a Humanized Mouse Model. Microorganisms 2023; 11:1131. [PMID: 37317106 PMCID: PMC10224461 DOI: 10.3390/microorganisms11051131] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/22/2023] [Accepted: 04/24/2023] [Indexed: 06/16/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is the leading basis for gastrointestinal morbidity and poses a significant risk for neurodevelopmental impairment (NDI) in preterm infants. Aberrant bacterial colonization preceding NEC contributes to the pathogenesis of NEC, and we have demonstrated that immature microbiota in preterm infants negatively impacts neurodevelopment and neurological outcomes. In this study, we tested the hypothesis that microbial communities before the onset of NEC drive NDI. Using our humanized gnotobiotic model in which human infant microbial samples were gavaged to pregnant germ-free C57BL/6J dams, we compared the effects of the microbiota from preterm infants who went on to develop NEC (MNEC) to the microbiota from healthy term infants (MTERM) on brain development and neurological outcomes in offspring mice. Immunohistochemical studies demonstrated that MNEC mice had significantly decreased occludin and ZO-1 expression compared to MTERM mice and increased ileal inflammation marked by the increased nuclear phospho-p65 of NFκB expression, revealing that microbial communities from patients who developed NEC had a negative effect on ileal barrier development and homeostasis. In open field and elevated plus maze tests, MNEC mice had worse mobility and were more anxious than MTERM mice. In cued fear conditioning tests, MNEC mice had worse contextual memory than MTERM mice. MRI revealed that MNEC mice had decreased myelination in major white and grey matter structures and lower fractional anisotropy values in white matter areas, demonstrating delayed brain maturation and organization. MNEC also altered the metabolic profiles, especially carnitine, phosphocholine, and bile acid analogs in the brain. Our data demonstrated numerous significant differences in gut maturity, brain metabolic profiles, brain maturation and organization, and behaviors between MTERM and MNEC mice. Our study suggests that the microbiome before the onset of NEC has negative impacts on brain development and neurological outcomes and can be a prospective target to improve long-term developmental outcomes.
Collapse
Affiliation(s)
- Jing Lu
- Department of Pediatrics, Division of Biological Sciences, Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA
| | | | - Lei Lu
- Department of Pediatrics, Division of Biological Sciences, Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Yueyue Yu
- Department of Pediatrics, Division of Biological Sciences, Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Michael S. Caplan
- Department of Pediatrics, NorthShore University HealthSystem, Evanston, IL 60202, USA
| | - Erika C. Claud
- Department of Pediatrics, Division of Biological Sciences, Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
24
|
Jovandaric MZ, Dugalic S, Babic S, Babovic IR, Milicevic S, Mihajlovic D, Culjic M, Zivanovic T, Trklja A, Markovic B, Plesinac V, Jestrovic Z, Medjo B, Raus M, Dugalic MG. Programming Factors of Neonatal Intestinal Dysbiosis as a Cause of Disease. Int J Mol Sci 2023; 24:5723. [PMID: 36982799 PMCID: PMC10058501 DOI: 10.3390/ijms24065723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/07/2023] [Accepted: 03/13/2023] [Indexed: 03/19/2023] Open
Abstract
The intestinal microbiota consists of trillions of bacteria, viruses, and fungi that achieve a perfect symbiosis with the host. They perform immunological, metabolic, and endocrine functions in the body. The microbiota is formed intrauterine. Dysbiosis is a microbiome disorder characterized by an imbalance in the composition of the microbiota, as well as changes in their functional and metabolic activities. The causes of dysbiosis include improper nutrition in pregnant women, hormone therapy, the use of drugs, especially antibiotics, and a lack of exposure to the mother's vaginal microbiota during natural birth. Changes in the intestinal microbiota are increasingly being identified in various diseases, starting in the early neonatal period into the adult period. Conclusions: In recent years, it has become more and more obvious that the components of the intestinal microbiota are crucial for the proper development of the immune system, and its disruption leads to disease.
Collapse
Affiliation(s)
- Miljana Z. Jovandaric
- Department of Neonatology, Clinic for Gynecology and Obstetrics, University Clinical Center of Serbia, 11000 Belgrade, Serbia
| | - Stefan Dugalic
- Department of Gynecology and Obstetrics, Clinic for Gynecology and Obstetrics, University Clinical Center of Serbia, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Sandra Babic
- Department of Gynecology and Obstetrics, Clinic for Gynecology and Obstetrics, University Clinical Center of Serbia, 11000 Belgrade, Serbia
| | - Ivana R. Babovic
- Department of Gynecology and Obstetrics, Clinic for Gynecology and Obstetrics, University Clinical Center of Serbia, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Srboljub Milicevic
- Department of Gynecology and Obstetrics, Clinic for Gynecology and Obstetrics, University Clinical Center of Serbia, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Dejan Mihajlovic
- Faculty of Medicine, University of Pristina Temporarily Settled in Kosovska Mitrovica, 38220, Serbia
| | - Miljan Culjic
- Department of Gynecology and Obstetrics, Clinic for Gynecology and Obstetrics, University Clinical Center of Serbia, 11000 Belgrade, Serbia
| | - Tamara Zivanovic
- Department of Gynecology and Obstetrics, Clinic for Gynecology and Obstetrics, University Clinical Center of Serbia, 11000 Belgrade, Serbia
| | - Aleksandar Trklja
- Department of Gynecology and Obstetrics, Clinic for Gynecology and Obstetrics, University Clinical Center of Serbia, 11000 Belgrade, Serbia
| | - Bogdan Markovic
- Department of Gynecology and Obstetrics, Clinic for Gynecology and Obstetrics, University Clinical Center of Serbia, 11000 Belgrade, Serbia
| | - Vera Plesinac
- Department of Gynecology and Obstetrics, Clinic for Gynecology and Obstetrics, University Clinical Center of Serbia, 11000 Belgrade, Serbia
| | - Zorica Jestrovic
- Department of Gynecology and Obstetrics, Clinic for Gynecology and Obstetrics, University Clinical Center of Serbia, 11000 Belgrade, Serbia
| | - Biljana Medjo
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Department Pediatrics and Neonatal Intensive Care, University Children’s Hospital, 11000 Belgrade, Serbia
| | - Misela Raus
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Department of Neonatology, University Children’s Hospital, 11000 Belgrade, Serbia
| | - Miroslava Gojnic Dugalic
- Department of Gynecology and Obstetrics, Clinic for Gynecology and Obstetrics, University Clinical Center of Serbia, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| |
Collapse
|
25
|
Neumann CJ, Mahnert A, Kumpitsch C, Kiu R, Dalby MJ, Kujawska M, Madl T, Kurath-Koller S, Urlesberger B, Resch B, Hall LJ, Moissl-Eichinger C. Clinical NEC prevention practices drive different microbiome profiles and functional responses in the preterm intestine. Nat Commun 2023; 14:1349. [PMID: 36906612 PMCID: PMC10008552 DOI: 10.1038/s41467-023-36825-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 02/13/2023] [Indexed: 03/13/2023] Open
Abstract
Preterm infants with very low birthweight are at serious risk for necrotizing enterocolitis. To functionally analyse the principles of three successful preventive NEC regimens, we characterize fecal samples of 55 infants (<1500 g, n = 383, female = 22) longitudinally (two weeks) with respect to gut microbiome profiles (bacteria, archaea, fungi, viruses; targeted 16S rRNA gene sequencing and shotgun metagenomics), microbial function, virulence factors, antibiotic resistances and metabolic profiles, including human milk oligosaccharides (HMOs) and short-chain fatty acids (German Registry of Clinical Trials, No.: DRKS00009290). Regimens including probiotic Bifidobacterium longum subsp. infantis NCDO 2203 supplementation affect microbiome development globally, pointing toward the genomic potential to convert HMOs. Engraftment of NCDO 2203 is associated with a substantial reduction of microbiome-associated antibiotic resistance as compared to regimens using probiotic Lactobacillus rhamnosus LCR 35 or no supplementation. Crucially, the beneficial effects of Bifidobacterium longum subsp. infantis NCDO 2203 supplementation depends on simultaneous feeding with HMOs. We demonstrate that preventive regimens have the highest impact on development and maturation of the gastrointestinal microbiome, enabling the establishment of a resilient microbial ecosystem that reduces pathogenic threats in at-risk preterm infants.
Collapse
Affiliation(s)
- Charlotte J Neumann
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine; Medical University of Graz, Graz, Styria, 8010, Austria
| | - Alexander Mahnert
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine; Medical University of Graz, Graz, Styria, 8010, Austria
| | - Christina Kumpitsch
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine; Medical University of Graz, Graz, Styria, 8010, Austria
| | - Raymond Kiu
- Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
| | - Matthew J Dalby
- Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
| | - Magdalena Kujawska
- Chair of Intestinal Microbiome, School of Life Sciences, ZIEL-Institute for Food & Health; Technical University of Munich, Freising, Bavaria, 85354, Germany
| | - Tobias Madl
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology & Biochemistry, Medical University of Graz, Graz, Styria, 8010, Austria
- BioTechMed, Graz, Styria, 8010, Austria
| | - Stefan Kurath-Koller
- Division of Paediatric Cardiology, Department of Paediatrics and Adolescent Medicine, Medical University of Graz, Graz, Styria, 8036, Austria
| | - Berndt Urlesberger
- Division of Neonatology; Department of Paediatrics and Adolescent Medicine, Medical University of Graz, Graz, Styria, 8036, Austria
- Research Unit for Neonatal Infectious Diseases and Epidemiology, Medical University of Graz, Graz, Styria, 8036, Austria
| | - Bernhard Resch
- Division of Neonatology; Department of Paediatrics and Adolescent Medicine, Medical University of Graz, Graz, Styria, 8036, Austria.
- Research Unit for Neonatal Infectious Diseases and Epidemiology, Medical University of Graz, Graz, Styria, 8036, Austria.
| | - Lindsay J Hall
- Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
- Chair of Intestinal Microbiome, School of Life Sciences, ZIEL-Institute for Food & Health; Technical University of Munich, Freising, Bavaria, 85354, Germany
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Christine Moissl-Eichinger
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine; Medical University of Graz, Graz, Styria, 8010, Austria.
- BioTechMed, Graz, Styria, 8010, Austria.
| |
Collapse
|
26
|
Giannella L, Grelloni C, Quintili D, Fiorelli A, Montironi R, Alia S, Delli Carpini G, Di Giuseppe J, Vignini A, Ciavattini A. Microbiome Changes in Pregnancy Disorders. Antioxidants (Basel) 2023; 12:463. [PMID: 36830021 PMCID: PMC9952029 DOI: 10.3390/antiox12020463] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023] Open
Abstract
The human microbiota comprises all microorganisms, such as bacteria, fungi, and viruses, found within a specific environment that live on our bodies and inside us. The last few years have witnessed an explosion of information related to the role of microbiota changes in health and disease. Even though the gut microbiota is considered the most important in maintaining our health, other regions of the human body, such as the oral cavity, lungs, vagina, and skin, possess their own microbiota. Recent work suggests a correlation between the microbiota present during pregnancy and pregnancy complications. The aim of our literature review was to provide a broad overview of this growing and important topic. We focused on the most significant changes in the microbiota in the four more common obstetric diseases affecting women's health. Thus, our attention will be focused on hypertensive disorders, gestational diabetes mellitus, preterm birth, and recurrent miscarriage. Pregnancy is a unique period in a woman's life since the body undergoes different adaptations to provide an optimal environment for fetal growth. Such changes also involve all the microorganisms, which vary in composition and quantity during the three trimesters of gestation. In addition, special attention will be devoted to the potential and fundamental advances in developing clinical applications to prevent and treat those disorders by modulating the microbiota to develop personalized therapies for disease prevention and tailored treatments.
Collapse
Affiliation(s)
- Luca Giannella
- Woman’s Health Sciences Department, Gynecologic Section, Polytechnic University of Marche, Via Filippo Corridoni, 16, 60123 Ancona, Italy
| | - Camilla Grelloni
- Woman’s Health Sciences Department, Gynecologic Section, Polytechnic University of Marche, Via Filippo Corridoni, 16, 60123 Ancona, Italy
| | - Dayana Quintili
- Woman’s Health Sciences Department, Gynecologic Section, Polytechnic University of Marche, Via Filippo Corridoni, 16, 60123 Ancona, Italy
| | - Alessia Fiorelli
- Woman’s Health Sciences Department, Gynecologic Section, Polytechnic University of Marche, Via Filippo Corridoni, 16, 60123 Ancona, Italy
| | - Ramona Montironi
- Woman’s Health Sciences Department, Gynecologic Section, Polytechnic University of Marche, Via Filippo Corridoni, 16, 60123 Ancona, Italy
| | - Sonila Alia
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Via Tronto 10/A, 60126 Ancona, Italy
| | - Giovanni Delli Carpini
- Woman’s Health Sciences Department, Gynecologic Section, Polytechnic University of Marche, Via Filippo Corridoni, 16, 60123 Ancona, Italy
| | - Jacopo Di Giuseppe
- Woman’s Health Sciences Department, Gynecologic Section, Polytechnic University of Marche, Via Filippo Corridoni, 16, 60123 Ancona, Italy
| | - Arianna Vignini
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Via Tronto 10/A, 60126 Ancona, Italy
| | - Andrea Ciavattini
- Woman’s Health Sciences Department, Gynecologic Section, Polytechnic University of Marche, Via Filippo Corridoni, 16, 60123 Ancona, Italy
| |
Collapse
|
27
|
Microbiota-immune-brain interactions: A lifespan perspective. Curr Opin Neurobiol 2023; 78:102652. [PMID: 36463579 DOI: 10.1016/j.conb.2022.102652] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/10/2022] [Accepted: 10/31/2022] [Indexed: 12/03/2022]
Abstract
There is growing appreciation of key roles of the gut microbiota in maintaining homeostasis and influencing brain and behaviour at critical windows across the lifespan. Mounting evidence suggests that communication between the gut and the brain could be the key to understanding multiple neuropsychiatric disorders, with the immune system coming to the forefront as an important mechanistic mediator. Throughout the lifespan, the immune system exchanges continuous reciprocal signals with the central nervous system. Intestinal microbial cues alter immune mediators with consequences for host neurophysiology and behaviour. Several factors challenge the gut microbiota composition, which in response release molecules with neuro- and immuno-active potential that are crucial for adequate neuro-immune interactions. In this review, multiple factors contributing to the upkeep of the fine balance between health and disease of these systems are discussed, and we elucidate the potential mechanistic implications for the gut microbiota inputs on host brain and behaviour across the lifespan.
Collapse
|
28
|
Lu J, Martin CR, Claud EC. Neurodevelopmental outcome of infants who develop necrotizing enterocolitis: The gut-brain axis. Semin Perinatol 2023; 47:151694. [PMID: 36572620 PMCID: PMC9974904 DOI: 10.1016/j.semperi.2022.151694] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Necrotizing enterocolitis (NEC) poses a significant risk for neurodevelopmental impairment in extremely preterm infants. The gut microbiota shapes the development of the gut, immune system, and the brain; and dysbiosis drive neonatal morbidities including NEC. In this chapter, we delineate a gut-brain axis linking gut microbiota to the adverse neurological outcomes in NEC patients. We propose that in NEC, immaturity of the microbiome along with aberrant gut microbiota-driven immaturity of the gut barrier and immune system can lead to effects including systemic inflammation and circulating microbial mediators. This nexus of gut microbiota-driven systemic effects further interacts with a likewise underdeveloped blood-brain barrier to regulate neuroinflammation and neurodevelopment. Targeting deviant gut-brain axis signaling presents an opportunity to improve the neurodevelopmental outcomes of NEC patients.
Collapse
Affiliation(s)
- Jing Lu
- Department of Pediatrics, Division of Biological Sciences, University of Chicago, Pritzker School of Medicine, Chicago, Illinois 60637, United States
| | - Camilia R Martin
- Department of Pediatrics, Division of Newborn Medicine, Weill Cornell Medicine, New York, New York 10021, United States
| | - Erika C Claud
- Department of Pediatrics, Division of Biological Sciences, University of Chicago, Pritzker School of Medicine, Chicago, Illinois 60637, United States.
| |
Collapse
|
29
|
Duque C, Chrisostomo DA, Souza ACA, de Almeida Braga GP, Dos Santos VR, Caiaffa KS, Pereira JA, de Oliveira WC, de Aguiar Ribeiro A, Parisotto TM. Understanding the Predictive Potential of the Oral Microbiome in the Development and Progression of Early Childhood Caries. Curr Pediatr Rev 2023; 19:121-138. [PMID: 35959611 DOI: 10.2174/1573396318666220811124848] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 03/24/2022] [Accepted: 04/22/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND Early childhood caries (ECC) is the most common chronic disease in young children and a public health problem worldwide. It is characterized by the presence of atypical and fast progressive caries lesions. The aggressive form of ECC, severe early childhood caries (S-ECC), can lead to the destruction of the whole crown of most of the deciduous teeth and cause pain and sepsis, affecting the child's quality of life. Although the multifactorial etiology of ECC is known, including social, environmental, behavioral, and genetic determinants, there is a consensus that this disease is driven by an imbalance between the oral microbiome and host, or dysbiosis, mediated by high sugar consumption and poor oral hygiene. Knowledge of the microbiome in healthy and caries status is crucial for risk monitoring, prevention, and development of therapies to revert dysbiosis and restore oral health. Molecular biology tools, including next-generation sequencing methods and proteomic approaches, have led to the discovery of new species and microbial biomarkers that could reveal potential risk profiles for the development of ECC and new targets for anti-caries therapies. This narrative review summarized some general aspects of ECC, such as definition, epidemiology, and etiology, the influence of oral microbiota in the development and progression of ECC based on the current evidence from genomics, transcriptomic, proteomic, and metabolomic studies and the effect of antimicrobial intervention on oral microbiota associated with ECC. CONCLUSION The evaluation of genetic and proteomic markers represents a promising approach to predict the risk of ECC before its clinical manifestation and plan efficient therapeutic interventions for ECC in its initial stages, avoiding irreversible dental cavitation.
Collapse
Affiliation(s)
- Cristiane Duque
- Department of Preventive and Restorative Dentistry, Araçatuba Dental School, State University of São Paulo (UNESP), Araçatuba, Brazil
| | - Daniela Alvim Chrisostomo
- Department of Preventive and Restorative Dentistry, Araçatuba Dental School, State University of São Paulo (UNESP), Araçatuba, Brazil
| | - Amanda Caselato Andolfatto Souza
- Department of Preventive and Restorative Dentistry, Araçatuba Dental School, State University of São Paulo (UNESP), Araçatuba, Brazil
| | - Gabriela Pacheco de Almeida Braga
- Department of Preventive and Restorative Dentistry, Araçatuba Dental School, State University of São Paulo (UNESP), Araçatuba, Brazil
| | - Vanessa Rodrigues Dos Santos
- Department of Preventive and Restorative Dentistry, Araçatuba Dental School, State University of São Paulo (UNESP), Araçatuba, Brazil
| | - Karina Sampaio Caiaffa
- Department of Preventive and Restorative Dentistry, Araçatuba Dental School, State University of São Paulo (UNESP), Araçatuba, Brazil
| | - Jesse Augusto Pereira
- Department of Preventive and Restorative Dentistry, Araçatuba Dental School, State University of São Paulo (UNESP), Araçatuba, Brazil
| | - Warlley Campos de Oliveira
- Department of Preventive and Restorative Dentistry, Araçatuba Dental School, State University of São Paulo (UNESP), Araçatuba, Brazil
| | - Apoena de Aguiar Ribeiro
- Division of Diagnostic Sciences, University of North Carolina at Chapel Hill - Adams School of Dentistry, Chapel Hill, North Carolina, United State
| | - Thaís Manzano Parisotto
- Laboratory of Clinical and Molecular Microbiology, São Francisco University, Bragança Paulista, Brazil
| |
Collapse
|
30
|
Lynch LE, Hair AB, Soni KG, Yang H, Gollins LA, Narvaez-Rivas M, Setchell KDR, Preidis GA. Cholestasis impairs gut microbiota development and bile salt hydrolase activity in preterm neonates. Gut Microbes 2023; 15:2183690. [PMID: 36843227 PMCID: PMC9980517 DOI: 10.1080/19490976.2023.2183690] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/28/2023] Open
Abstract
Cholestasis refers to impaired bile flow from the liver to the intestine. In neonates, cholestasis causes poor growth and may progress to liver failure and death. Normal bile flow requires an intact liver-gut-microbiome axis, whereby liver-derived primary bile acids are transformed into secondary bile acids. Microbial bile salt hydrolase (BSH) enzymes are responsible for the first step, deconjugating glycine- and taurine-conjugated primary bile acids. Cholestatic neonates often are treated with the potent choleretic bile acid ursodeoxycholic acid (UDCA), although interactions between UDCA, gut microbes, and other bile acids are poorly understood. To gain insight into how the liver-gut-microbiome axis develops in extreme prematurity and how cholestasis alters this maturation, we conducted a nested case-control study collecting 124 stool samples longitudinally from 24 preterm infants born at mean 27.2 ± 1.8 weeks gestation and 946 ± 249.6 g, half of whom developed physiologic cholestasis. Samples were analyzed by whole metagenomic sequencing, in vitro BSH enzyme activity assays optimized for low biomass fecal samples, and quantitative mass spectrometry to measure the bile acid metabolome. In extremely preterm neonates, acquisition of the secondary bile acid biosynthesis pathway and BSH genes carried by Clostridium perfringens are the most prominent features of early microbiome development. Cholestasis interrupts this developmental pattern. BSH gene abundance and enzyme activity are profoundly reduced in cholestatic neonates, resulting in decreased quantities of unconjugated bile acids. UDCA restores total fecal bile acid levels in cholestatic neonates, but this is due to a 522-fold increase in fecal UDCA. A majority of bile acids in early development are atypical positional and stereo-isomers of bile acids. We report novel associations linking isomeric bile acids and BSH activity to neonatal growth trajectories. These data highlight deconjugation of bile acids as a key microbial function that is acquired in early neonatal development and impaired by cholestasis.
Collapse
Affiliation(s)
- Lauren E. Lynch
- Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| | - Amy B. Hair
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA,CONTACT Amy B. Hair Division of Neonatology, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, 6621 Fannin Street, Suite A5590, Houston, TX77030, USA
| | - Krishnakant G. Soni
- Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| | - Heeju Yang
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| | - Laura A. Gollins
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| | - Monica Narvaez-Rivas
- Division of Pathology and Laboratory Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Kenneth D. R. Setchell
- Division of Pathology and Laboratory Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Geoffrey A. Preidis
- Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA,Geoffrey A. Preidis Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, 1102 Bates Avenue, Feigin Tower Suite 860, Houston, TX77030, USA
| |
Collapse
|
31
|
Characteristics of Gut Microbiota in Small for Gestational Age Infants with Very Low Birth Weight. Nutrients 2022; 14:nu14235158. [PMID: 36501188 PMCID: PMC9738608 DOI: 10.3390/nu14235158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/22/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Small for gestational age (SGA) birth is associated with high rates of mortality and morbidity in preterm infants. The aim of this preliminary observational study was to investigate the difference in gut microbiota between SGA and appropriate for gestational age (AGA) preterm infants with very low birth weight (VLBW). We included 20 VLBW preterm infants (SGA, n = 10; AGA, n = 10) in this study. Stool samples were collected on days 7, 14, and 30 after birth. We performed 16S ribosomal DNA sequencing to compare microbiota composition between both groups. The SGA group exhibited a lower abundance of Klebsiella on day 14 (SGA, 0.57%; AGA, 7.42%; p = 0.037). On day 30, the SGA group exhibited a lower abundance of Klebsiella (SGA 3.76% vs. AGA 16.05%; p = 0.07) and Enterobacter (SGA 5.09% vs. AGA 27.25%; p = 0.011) than the AGA group. Beta diversity demonstrated a separation of the bacterial community structure between both groups on day 30 (p = 0.019). The present study revealed that a distinct gut microbiota profile gradually develops in SGA preterm infants with VLBW during the early days of life. The role of changes in gut microbiota structure warrants further investigation.
Collapse
|
32
|
Obiero CW, Gumbi W, Mwakio S, Mwangudzah H, Seale AC, Taniuchi M, Liu J, Houpt E, Berkley JA. Detection of pathogens associated with early-onset neonatal sepsis in cord blood at birth using quantitative PCR. Wellcome Open Res 2022; 7:3. [PMID: 35600002 PMCID: PMC9114825 DOI: 10.12688/wellcomeopenres.17386.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2022] [Indexed: 11/10/2022] Open
Abstract
Background: Early onset neonatal sepsis (EONS) typically begins prior to, during or soon after birth and may be rapidly fatal. There is paucity of data on the aetiology of EONS in sub-Saharan Africa due to limited diagnostic capacity in this region, despite the associated significant mortality and long-term neurological impairment. Methods: We compared pathogens detected in cord blood samples between neonates admitted to hospital with possible serious bacterial infection (pSBI) in the first 48 hours of life (cases) and neonates remaining well (controls). Cord blood was systematically collected at Kilifi County Hospital (KCH) from 2011-2016, and later tested for 21 bacterial, viral and protozoal targets using multiplex PCR via TaqMan Array Cards (TAC). Results: Among 603 cases (101 [17%] of whom died), 179 (30%) tested positive for ≥1 target and 37 (6.1%) tested positive for multiple targets. Klebsiella oxytoca, Escherichia coli/Shigella spp., Pseudomonas aeruginosa, and Streptococcus pyogenes were commonest. Among 300 controls, 79 (26%) tested positive for ≥1 target, 11 (3.7%) were positive for multiple targets, and K. oxytoca and P. aeruginosa were most common. Cumulative odds ratios across controls: cases (survived): cases (died) were E. coli/Shigella spp. 2.6 (95%CI 1.6-4.4); E. faecalis 4.0 (95%CI 1.1-15); S. agalactiae 4.5 (95%CI 1.6-13); Ureaplasma spp. 2.9 (95%CI 1.3-6.4); Enterovirus 9.1 (95%CI 2.3-37); and Plasmodium spp. 2.9 (95%CI 1.4-6.2). Excluding K. oxytoca and P. aeruginosa as likely contaminants, aetiology was attributed in 9.4% (95%CI 5.1-13) cases using TAC. Leading pathogen attributions by TAC were E. coli/Shigella spp. (3.5% (95%CI 1.7-5.3)) and Ureaplasma spp. (1.7% (95%CI 0.5-3.0)). Conclusions: Cord blood sample may be useful in describing EONS pathogens at birth, but more specific tests are needed for individual diagnosis. Careful sampling of cord blood using aseptic techniques is crucial to minimize contamination. In addition to culturable bacteria, Ureaplasma and Enterovirus were causes of EONS.
Collapse
Affiliation(s)
- Christina W. Obiero
- Clinical research, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Global health, Academic Medical Center of the University of Amsterdam, Amsterdam, The Netherlands
| | - Wilson Gumbi
- Bioscience department, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Stella Mwakio
- Clinical research, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Hope Mwangudzah
- Clinical research, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Anna C. Seale
- Clinical research, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
- College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Mami Taniuchi
- Division of Infectious Diseases and International Health, University of Virginia, Virginia, USA
| | - Jie Liu
- Division of Infectious Diseases and International Health, University of Virginia, Virginia, USA
| | - Eric Houpt
- Division of Infectious Diseases and International Health, University of Virginia, Virginia, USA
| | - James A. Berkley
- Clinical research, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine, University of Oxford, Oxford, UK
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi, Kenya
| |
Collapse
|
33
|
Vongbhavit K, Salinero LK, Kalanetra KM, Masarweh C, Yu A, Taft DH, Mills DA, Underwood MA. A comparison of bacterial colonization between nasogastric and orogastric enteral feeding tubes in infants in the neonatal intensive care unit. J Perinatol 2022; 42:1446-1452. [PMID: 35840710 PMCID: PMC9616717 DOI: 10.1038/s41372-022-01452-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 06/03/2022] [Accepted: 06/27/2022] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Feeding tubes harbor microbial contaminants; studies to date have not explored differences between orogastric (OG) and nasogastric (NG) tube biofilms. We sought to extend a previous analysis by comparing bacterial colonization by location (OG v NG) and by evaluating clinical factors that may affect tube bacterial populations. STUDY DESIGN The pharyngeal segments of 41 infant feeding tubes (14 OG and 27 NG) from 41 infants were analyzed by next generation 16 S rRNA sequencing on the MiSeq platform. RESULTS At the phylum level, Proteobacteria had the highest relative abundance of both OG and NG tubes. At the genus/species level, nine taxa differed significantly between OG and NG tubes. Alpha and beta diversity analyses showed significant differences between OG and NG tubes with relatively little contribution from clinical factors. CONCLUSION The route of feeding tube insertion (oral vs nasal) had a greater impact on bacterial colonization than the assessed clinical factors.
Collapse
Affiliation(s)
- Kannikar Vongbhavit
- Department of Pediatrics, HRH Princess Maha Chakri Sirindhorn Medical Center, Srinakharinwirot University, Nakornayok, Thailand
- Division of Neonatology, Department of Pediatrics, University of California Davis, Sacramento, CA, USA
| | - Lauren K Salinero
- Department of Food Science and Technology, University of California Davis, Davis, CA, USA
| | - Karen M Kalanetra
- Department of Food Science and Technology, University of California Davis, Davis, CA, USA
| | - Chad Masarweh
- Department of Food Science and Technology, University of California Davis, Davis, CA, USA
| | - Alice Yu
- Department of Food Science and Technology, University of California Davis, Davis, CA, USA
| | - Diana H Taft
- Department of Food Science and Technology, University of California Davis, Davis, CA, USA
| | - David A Mills
- Department of Food Science and Technology, University of California Davis, Davis, CA, USA
| | - Mark A Underwood
- Division of Neonatology, Department of Pediatrics, University of California Davis, Sacramento, CA, USA.
| |
Collapse
|
34
|
Otten K, Keller L, Puiu AA, Herpertz-Dahlmann B, Seitz J, Kohn N, Edgar JC, Wagels L, Konrad K. Pre- and postnatal antibiotic exposure and risk of developing attention deficit hyperactivity disorder-A systematic review and meta-analysis combining evidence from human and animal studies. Neurosci Biobehav Rev 2022; 140:104776. [PMID: 35842009 DOI: 10.1016/j.neubiorev.2022.104776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 07/10/2022] [Indexed: 11/26/2022]
Abstract
This study investigated the effects of early antibiotic exposure on ADHD risk by (1) integrating meta-analytical evidence from human observational studies examining the association between prenatal or early postnatal antibiotic exposure on the risk of developing ADHD; and (2) reviewing evidence from experimental animal studies on the effects of early antibiotic exposure on behavior. Sixteen human studies and five rodent studies were reviewed. A quantitative meta-analysis with 10 human studies indicated an increased risk for ADHD after prenatal antibiotic exposure (summary effect estimate Hazard Ratio (HR) 1.23, 95% CI 1.09-1.38; N = 2,398,475 subjects) but not after postnatal exposure within the first two years of life (summary effect estimate HR 1.12, 95% CI 0.95-1.32; N = 1,863,867 subjects). The rodent literature suggested that peri-natal antibiotic exposure has effects on social behavior, anxiety and aggression, alongside changes in gut microbial composition. Human and rodent findings thus suggest prenatal antibiotic exposure as a possible risk factor for ADHD, and suggest that an early disruption of the gut microbiome by antibiotics may interfere with neurodevelopment.
Collapse
Affiliation(s)
- Katharina Otten
- Child Neuropsychology Section, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany; Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine, RWTH Aachen University, Aachen, Germany.
| | - Lara Keller
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Faculty of Medicine, RWTH University Aachen, Aachen, Germany
| | - Andrei A Puiu
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Beate Herpertz-Dahlmann
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Faculty of Medicine, RWTH University Aachen, Aachen, Germany
| | - Jochen Seitz
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Faculty of Medicine, RWTH University Aachen, Aachen, Germany
| | - Nils Kohn
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - J Christopher Edgar
- Lurie Family Foundations MEG Imaging Center, Department of Radiology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lisa Wagels
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine, RWTH Aachen University, Aachen, Germany; Institute of Neuroscience and Medicine 10, Research Centre Jülich, Germany
| | - Kerstin Konrad
- Child Neuropsychology Section, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany; JARA-Brain Institute II, Molecular Neuroscience and Neuroimaging (INM-11), RWTH Aachen & Research Centre Jülich, 52428 Jülich, Germany
| |
Collapse
|
35
|
Ting JY, Yoon EW, Fajardo CA, Daboval T, Bertelle V, Shah PS. Antimicrobial utilization in very-low-birth-weight infants: association with probiotic use. J Perinatol 2022; 42:947-952. [PMID: 35399098 DOI: 10.1038/s41372-022-01382-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 03/12/2022] [Accepted: 03/22/2022] [Indexed: 11/09/2022]
Abstract
OBJECTIVE To examine the association between probiotic use and antimicrobial utilization. STUDY DESIGN We retrospectively evaluated very-low-birth-weight (VLBW) infants admitted to tertiary neonatal intensive care units in Canada between 2014 and 2019. Our outcome was antimicrobial utilization rate (AUR) defined as number of days of antimicrobial exposure per 1000 patient-days. RESULT Of 16,223 eligible infants, 7279 (45%) received probiotics. Probiotic use rate increased from 10% in 2014 to 68% in 2019. The AUR was significantly lower in infants who received probiotics vs those who did not (107 vs 129 per 1000 patient-days, aRR = 0.89, 95% CI [0.81, 0.98]). Among 13,305 infants without culture-proven sepsis or necrotizing enterocolitis ≥Stage 2, 5931 (45%) received probiotics. Median AUR was significantly lower in the probiotic vs the no-probiotic group (78 vs 97 per 1000 patient-days, aRR = 0.85, 95% CI [0.74, 0.97]). CONCLUSION Probiotic use was associated with a significant reduction in AUR among VLBW infants.
Collapse
Affiliation(s)
- Joseph Y Ting
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada.,Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Eugene W Yoon
- Maternal-infant Care Research Centre, Mount Sinai Hospital, Toronto, ON, Canada
| | - Carlos A Fajardo
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
| | - Thierry Daboval
- Department of Pediatrics, University of Ottawa, Ottawa, ON, Canada
| | - Valérie Bertelle
- Department of Pediatrics, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Prakesh S Shah
- Maternal-infant Care Research Centre, Mount Sinai Hospital, Toronto, ON, Canada. .,Department of Paediatrics, University of Toronto, Toronto, ON, Canada.
| | | |
Collapse
|
36
|
Abstract
Extremely preterm infants are particularly vulnerable to systemic infections secondary to their immature immune defenses, prolonged hospitalizations, delays in enteral feeding, early antibiotic exposure, and need for life-sustaining invasive interventions. There have been several evidence-based practices for infection prevention in this population, such as human milk feedings, utilization of "bundle checklists" and decolonization of pathogenic organisms. Other practices, such as the use of probiotics, human milk-derived fortifiers, and antifungal prophylaxis are more controversial and require further investigation regarding the risks and benefits of such interventions. This chapter examines the susceptibility of the preterm newborn infant to invasive infections and describes several strategies for infection prevention, along with the associated limitations of such practices. It also addresses the various gaps in our understanding of preventing infections in this population, and the need for additional large multi-center randomized controlled trials. Additionally, the role of the SARs-CoV-2 global pandemic and associated strategies for infection prevention in the NICU are discussed.
Collapse
|
37
|
Park HW. The Use of Probiotics in Preterm Infants. NEONATAL MEDICINE 2022. [DOI: 10.5385/nm.2022.29.2.68] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Probiotics are live microorganisms that positively affect host health by altering the composition of the host microbiota. Gastrointestinal dysbiosis refers to adverse alterations of the intestinal flora and is associated with several diseases, including necrotizing enterocolitis, late-onset sepsis in preterm infants as well as atopic disease, colic, diabetes, and diarrhea in term infants. The risk factors for gastrointestinal dysbiosis are preterm birth, cesarean section delivery, and formula feeding, in contrast to term birth infants, vaginal delivery and breast milk feeding. Probiotics have been used to restore synbiosis in infants with gastrointestinal dysbiosis. Probiotics inhibit colonization of pathogenic bacteria in the gastrointestinal tract, thereby improving the barrier function of the gastrointestinal tract, and the immune function. In preterm infants, probiotics reduce mortality as well as rates of necrotizing enterocolitis and late-onset sepsis. The combined use of probiotics such as <i>Lactobacillus</i> and <i>Bifidobacterium</i> and the combination of probiotics with prebiotics yield better outcomes in the prevention of necrotizing enterocolitis than those achieved with a single pro- or prebiotic strain. However, the routine use of probiotics has been hindered by the lack of pharmaceutical-quality products, and a definite effect has yet to be demonstrated in preterm infants with a birth weight <1,000 g. Therefore, to reduce the risk of necrotizing enterocolitis in preterm infants, probiotics should be provided along with breast milk and other strategies aimed at preventing gastrointestinal dysbiosis.
Collapse
|
38
|
Emodin Ameliorates Intestinal Dysfunction by Maintaining Intestinal Barrier Integrity and Modulating the Microbiota in Septic Mice. Mediators Inflamm 2022; 2022:5026103. [PMID: 35677734 PMCID: PMC9168211 DOI: 10.1155/2022/5026103] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 03/26/2022] [Accepted: 04/21/2022] [Indexed: 12/12/2022] Open
Abstract
Sepsis-induced inflammatory response leads to intestinal damage and secondary bacterial translocation, causing systemic infections and eventually death. Emodin is a natural anthraquinone derivative in many plants with promising bioactivities. However, the effects and mechanisms of emodin on sepsis-induced intestinal dysfunctions have not been well clarified yet. We found that emodin treatment suppressed the inflammatory response in the intestines of septic mice. Intestinal barrier function was also improved by emodin through enhancing ZO-1 and occludin expression, which prevented the secondary translocation of Escherichia coli. By proteome microarray investigation, JNK2 was identified as a direct target of emodin. In vitro study also showed that emodin inhibited LPS-induced inflammatory response in intestinal epithelial cells. Nuclear factors including NF-κB and AP-1 were further identified as downstream effectors of JNK2. Bioinformatic analysis based on 16s rRNA gene sequencing illustrated that emodin treatment significantly increased the alpha- and beta-diversity of gut microbiota in septic mice. Moreover, data according to functional prediction showed that emodin decreased the abundance of potential pathogenic bacteria in gut. Our findings have shown that emodin treatment prevented inflammatory induced barrier dysfunction and decreased the potential pathogenicity of lumen bacteria, reducing the hazard of lumen bacterial translocation during sepsis.
Collapse
|
39
|
Obiero CW, Gumbi W, Mwakio S, Mwangudzah H, Seale AC, Taniuchi M, Liu J, Houpt E, Berkley JA. Detection of pathogens associated with early-onset neonatal sepsis in cord blood at birth using quantitative PCR. Wellcome Open Res 2022; 7:3. [PMID: 35600002 PMCID: PMC9114825 DOI: 10.12688/wellcomeopenres.17386.2] [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] [Accepted: 05/04/2022] [Indexed: 11/20/2022] Open
Abstract
Background: Early onset neonatal sepsis (EONS) typically begins prior to, during or soon after birth and may be rapidly fatal. There is paucity of data on the aetiology of EONS in sub-Saharan Africa due to limited diagnostic capacity in this region, despite the associated significant mortality and long-term neurological impairment. Methods: We compared pathogens detected in cord blood samples between neonates admitted to hospital with possible serious bacterial infection (pSBI) in the first 48 hours of life (cases) and neonates remaining well (controls). Cord blood was systematically collected at Kilifi County Hospital (KCH) from 2011-2016, and later tested for 21 bacterial, viral and protozoal targets using multiplex PCR via TaqMan Array Cards (TAC). Results: Among 603 cases (101 [17%] of whom died), 179 (30%) tested positive for ≥1 target and 37 (6.1%) tested positive for multiple targets. Klebsiella oxytoca, Escherichia coli/Shigella spp., Pseudomonas aeruginosa, and Streptococcus pyogenes were commonest. Among 300 controls, 79 (26%) tested positive for ≥1 target, 11 (3.7%) were positive for multiple targets, and K. oxytoca and P. aeruginosa were most common. Cumulative odds ratios across controls: cases (survived): cases (died) were E. coli/Shigella spp. 2.6 (95%CI 1.6-4.4); E. faecalis 4.0 (95%CI 1.1-15); S. agalactiae 4.5 (95%CI 1.6-13); Ureaplasma spp. 2.9 (95%CI 1.3-6.4); Enterovirus 9.1 (95%CI 2.3-37); and Plasmodium spp. 2.9 (95%CI 1.4-6.2). Excluding K. oxytoca and P. aeruginosa as likely contaminants, aetiology was attributed in 9.4% (95%CI 5.1-13) cases using TAC. Leading pathogen attributions by TAC were E. coli/Shigella spp. (3.5% (95%CI 1.7-5.3)) and Ureaplasma spp. (1.7% (95%CI 0.5-3.0)). Conclusions: Cord blood sample may be useful in describing EONS pathogens at birth, but more specific tests are needed for individual diagnosis. Careful sampling of cord blood using aseptic techniques is crucial to minimize contamination. In addition to culturable bacteria, Ureaplasma and Enterovirus were causes of EONS.
Collapse
Affiliation(s)
- Christina W. Obiero
- Clinical research, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Global health, Academic Medical Center of the University of Amsterdam, Amsterdam, The Netherlands
| | - Wilson Gumbi
- Bioscience department, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Stella Mwakio
- Clinical research, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Hope Mwangudzah
- Clinical research, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Anna C. Seale
- Clinical research, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
- College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Mami Taniuchi
- Division of Infectious Diseases and International Health, University of Virginia, Virginia, USA
| | - Jie Liu
- Division of Infectious Diseases and International Health, University of Virginia, Virginia, USA
| | - Eric Houpt
- Division of Infectious Diseases and International Health, University of Virginia, Virginia, USA
| | - James A. Berkley
- Clinical research, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine, University of Oxford, Oxford, UK
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi, Kenya
| |
Collapse
|
40
|
The impact of maternal asthma on the preterm infants' gut metabolome and microbiome (MAP study). Sci Rep 2022; 12:6437. [PMID: 35440708 PMCID: PMC9018729 DOI: 10.1038/s41598-022-10276-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 03/30/2022] [Indexed: 11/20/2022] Open
Abstract
Preterm infants are at a greater risk for the development of asthma and atopic disease, which can lead to lifelong negative health consequences. This may be due, in part, to alterations that occur in the gut microbiome and metabolome during their stay in the Neonatal Intensive Care Unit (NICU). To explore the differential roles of family history (i.e., predisposition due to maternal asthma diagnosis) and hospital-related environmental and clinical factors that alter microbial exposures early in life, we considered a unique cohort of preterm infants born ≤ 34 weeks gestational age from two local level III NICUs, as part of the MAP (Microbiome, Atopic disease, and Prematurity) Study. From MAP participants, we chose a sub-cohort of infants whose mothers had a history of asthma and matched gestational age and sex to infants of mothers without a history of asthma diagnosis (control). We performed a prospective, paired metagenomic and metabolomic analysis of stool and milk feed samples collected at birth, 2 weeks, and 6 weeks postnatal age. Although there were clinical factors associated with shifts in the diversity and composition of stool-associated bacterial communities, maternal asthma diagnosis did not play an observable role in shaping the infant gut microbiome during the study period. There were significant differences, however, in the metabolite profile between the maternal asthma and control groups at 6 weeks postnatal age. The most notable changes occurred in the linoleic acid spectral network, which plays a role in inflammatory and immune pathways, suggesting early metabolomic changes in the gut of preterm infants born to mothers with a history of asthma. Our pilot study suggests that a history of maternal asthma alters a preterm infants’ metabolomic pathways in the gut, as early as the first 6 weeks of life.
Collapse
|
41
|
Aguilar-Lopez M, Wetzel C, MacDonald A, Ho TTB, Donovan SM. Metagenomic profile of the fecal microbiome of preterm infants consuming mother's own milk with bovine milk-based fortifier or infant formula: a cross-sectional study. Am J Clin Nutr 2022; 116:435-445. [PMID: 35383822 PMCID: PMC9638768 DOI: 10.1093/ajcn/nqac081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/29/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Preterm (PT) infants harbor a different gut microbiome than full-term infants. Multiple factors affect gut microbial colonization of PT infants, including low gestational age, high rates of Cesarean section, exposure to antibiotics, and diet. Human milk, whether it's mother's own milk (MOM) or donor human milk, is the preferred feeding mode for PT infants but needs to be fortified to achieve adequate nutrient content. Infant formulas are introduced at later stages if human milk is insufficient or unavailable. How these dietary exposures affect the gut microbiome of PT infants is poorly understood. OBJECTIVES The goal of this study was to evaluate the metagenomic potential of the fecal microbiome of PT infants consuming MOM with bovine milk-based fortifier compared with PT formula alone. METHODS Forty-two stool samples, from 27 infants consuming MOM or formula (21 samples in each group) were included. Twelve infants had repeated sampling (2-3 samples). Shotgun genomic DNA sequencing was performed and analyzed using MetaPhlAn and HUMAnN2. Multivariate regression analysis, adjusting by the repeated sampling, was used to identify the features that differed between PT infants consuming MOM compared with formula. RESULTS The primary function of the fecal microbiome of PT infants was characterized by a high abundance of biosynthesis pathways. A set of core features was identified; these belonged to pathways for amino acid metabolism and vitamin K-2 biosynthesis. Five pathways significantly differed between the MOM and formula group. Pathways for fatty acid and carbohydrate degradation were significantly higher in the MOM group. Taxonomically, members of the phylum Actinobacteria and the genus Bifidobacterium were higher in PT infants exposed to MOM. CONCLUSIONS This study provides insight into the influence of feeding MOM compared with infant formula on the structure and function of the fecal microbiome of PT infants.
Collapse
Affiliation(s)
- Miriam Aguilar-Lopez
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | | | | | - Thao T B Ho
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | | |
Collapse
|
42
|
Letouzey M, Lorthe E, Marchand-Martin L, Kayem G, Charlier C, Butin M, Mitha A, Kaminski M, Benhammou V, Ancel PY, Boileau P, Foix-L'Hélias L. Early Antibiotic Exposure and Adverse Outcomes in Very Preterm Infants at Low Risk of Early-Onset Sepsis: The EPIPAGE-2 Cohort Study. J Pediatr 2022; 243:91-98.e4. [PMID: 34942178 DOI: 10.1016/j.jpeds.2021.11.075] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 10/06/2021] [Accepted: 11/21/2021] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To assess the association between early empirical antibiotics and neonatal adverse outcomes in very preterm infants without risk factors for early-onset sepsis (EOS). STUDY DESIGN This is a secondary analysis of the EPIPAGE-2 study, a prospective national population-based cohort that included all liveborn infants at 22-31 completed weeks of gestation in France in 2011. Infants at high risk of EOS (ie, born after preterm labor or preterm premature rupture of membranes or from a mother who had clinical chorioamnionitis or had received antibiotics during the last 72 hours) were excluded. Early antibiotic exposure was defined as antibiotic therapy started at day 0 or day 1 of life, irrespective of the duration and type of antibiotics. We compared treated and untreated patients using inverse probability of treatment weighting based on estimated propensity scores. RESULTS Among 648 very preterm infants at low risk of EOS, 173 (26.2%) had received early antibiotic treatment. Early antibiotic exposure was not associated with death or late-onset sepsis or necrotizing enterocolitis (OR, 1.04; 95% CI, 0.72-1.50); however, it was associated with higher odds of severe cerebral lesions (OR, 2.71; 95% CI, 1.25-5.86) and moderate-severe bronchopulmonary dysplasia (BPD) (OR, 2.30; 95% CI, 1.21-4.38). CONCLUSIONS Early empirical antibiotic therapy administrated in very preterm infants at low risk of EOS was associated with a higher risk of severe cerebral lesions and moderate-severe BPD.
Collapse
Affiliation(s)
- Mathilde Letouzey
- Université de Paris, Epidemiology and Statistics Research Center/CRESS, Obstetrical Perinatal and Pediatric Epidemiology Research Team (Epopé), Inserm, INRA, F-75004 Paris, France; Department of Neonatal Pediatrics, Poissy Saint Germain Hospital, Poissy, France
| | - Elsa Lorthe
- Université de Paris, Epidemiology and Statistics Research Center/CRESS, Obstetrical Perinatal and Pediatric Epidemiology Research Team (Epopé), Inserm, INRA, F-75004 Paris, France; Unit of Population Epidemiology, Department of Primary Care Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Laetitia Marchand-Martin
- Université de Paris, Epidemiology and Statistics Research Center/CRESS, Obstetrical Perinatal and Pediatric Epidemiology Research Team (Epopé), Inserm, INRA, F-75004 Paris, France
| | - Gilles Kayem
- Université de Paris, Epidemiology and Statistics Research Center/CRESS, Obstetrical Perinatal and Pediatric Epidemiology Research Team (Epopé), Inserm, INRA, F-75004 Paris, France; Department of Gynecology and Obstetrics, Armand Trousseau Hospital, APHP, Paris Sorbonne University, Paris, France
| | - Caroline Charlier
- Division of Infectious Diseases and Tropical Medicine, Hôpital Université Necker-Enfants Malades, Université de Paris, APHP, Paris, France; Biology of Infection Unit, Institut Pasteur, French National Reference Center and WHO Collaborating Center for Listeria, Inserm U1117, Paris, France
| | - Marine Butin
- Department of Neonatal Pediatrics, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Lyon, France
| | - Ayoub Mitha
- Department of Neonatal Medicine, Jeanne de Flandre Hospital, CHRU Lille, Lille, France
| | - Monique Kaminski
- Université de Paris, Epidemiology and Statistics Research Center/CRESS, Obstetrical Perinatal and Pediatric Epidemiology Research Team (Epopé), Inserm, INRA, F-75004 Paris, France
| | - Valerie Benhammou
- Université de Paris, Epidemiology and Statistics Research Center/CRESS, Obstetrical Perinatal and Pediatric Epidemiology Research Team (Epopé), Inserm, INRA, F-75004 Paris, France
| | - Pierre-Yves Ancel
- Université de Paris, Epidemiology and Statistics Research Center/CRESS, Obstetrical Perinatal and Pediatric Epidemiology Research Team (Epopé), Inserm, INRA, F-75004 Paris, France; URC-CIC P1419, Cochin Hotel-Dieu Hospital, APHP, Paris, France
| | - Pascal Boileau
- Department of Neonatal Pediatrics, Poissy Saint Germain Hospital, Poissy, France; Université Paris-Saclay, UVSQ, UFR Simone Veil Santé, Montigny Le Bretonneux, France
| | - Laurence Foix-L'Hélias
- Université de Paris, Epidemiology and Statistics Research Center/CRESS, Obstetrical Perinatal and Pediatric Epidemiology Research Team (Epopé), Inserm, INRA, F-75004 Paris, France; Department of Neonatal Pediatrics, Armand Trousseau Hospital, APHP, Paris Sorbonne University, Paris, France
| | | |
Collapse
|
43
|
Lu J, Lu L, Yu Y, Oliphant K, Drobyshevsky A, Claud EC. Early preterm infant microbiome impacts adult learning. Sci Rep 2022; 12:3310. [PMID: 35228616 PMCID: PMC8885646 DOI: 10.1038/s41598-022-07245-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 02/10/2022] [Indexed: 12/14/2022] Open
Abstract
Interventions to mitigate long-term neurodevelopmental deficits such as memory and learning impairment in preterm infants are warranted. Manipulation of the gut microbiome affects host behaviors. In this study we determined whether early maturation of the infant microbiome is associated with neurodevelopment outcomes. Germ free mice colonized at birth with human preterm infant microbiomes from infants of advancing post menstrual age (PMA) demonstrated an increase in bacterial diversity and a shift in dominance of taxa mimicking the human preterm microbiome development pattern. These characteristics along with changes in a number of metabolites as the microbiome matured influenced associative learning and memory but not locomotor ability, anxiety-like behaviors, or social interaction in adult mice. As a regulator of learning and memory, brain glial cell-derived neurotrophic factor increased with advancing PMA and was also associated with better performance in associative learning and memory in adult mice. We conclude that maturation of the microbiome in early life of preterm infants primes adult associative memory and learning ability. Our findings suggest a critical window of early intervention to affect maturation of the preterm infant microbiome and ultimately improve neurodevelopmental outcomes.
Collapse
Affiliation(s)
- Jing Lu
- Department of Pediatrics, Pritzker School of Medicine/Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Lei Lu
- Department of Pediatrics, Pritzker School of Medicine/Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Yueyue Yu
- Department of Pediatrics, Pritzker School of Medicine/Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Kaitlyn Oliphant
- Department of Pediatrics, Pritzker School of Medicine/Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Alexander Drobyshevsky
- Department of Pediatrics, NorthShore University HealthSystem Research Institute, Evanston, IL, 60202, USA
| | - Erika C Claud
- Department of Pediatrics, Pritzker School of Medicine/Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
44
|
Liu XC, Sun Q, Ji YC, Fu LZ, Wang ZL, He Y, Li LQ. Differences in the Gut Microbiota Composition and Metabolites Associated With Feeding Intolerance in VLBW Infants With a Gestational Age of ≤ 30 Weeks: A Pilot Study. Front Cell Infect Microbiol 2022; 12:726322. [PMID: 35252022 PMCID: PMC8891543 DOI: 10.3389/fcimb.2022.726322] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 01/27/2022] [Indexed: 12/12/2022] Open
Abstract
Objective To explore the main variations in gut microbiota compositions, short-chain fatty acids (SCFAs) concentrations and autoinducer-2 (AI-2) levels in very-low-birth-weight (VLBW) infants with feeding intolerance (FI). Methods Twenty-seven VLBW infants with gestational ages of ≤30 weeks were divided into the FI group (n=14) and feeding tolerance (FT) group (n=13). The gut microbiota composition and SCFAs concentrations and AI-2 levels in feces were detected at 2 and 4 weeks after birth. Results There was no difference in alpha diversity between the two groups at 2 and 4 weeks after birth (P>0.05). Although the Chao index decreased (P<0.05), there was no difference in the Shannon index from 2 weeks to 4 weeks in either the FI or FT group (P>0.05). Additionally, there was no difference in beta diversity between the FI and FT groups at 2 weeks (P>0.05), but there was a significant difference in beta diversity between the two groups at 4 weeks (P<0.05) and a large difference from 2 weeks to 4 weeks in both the FI and FT groups (P<0.05). Furthermore, the composition of the microbiota at 4 weeks was significantly different from that at 2 weeks in the FI group (P<0.05). The Veillonella abundance was lower at 4 weeks in the FI group (P<0.05), but there were no differences in the compositions of the other main microbes between the two groups (P>0.05). Proteobacteria and Firmicutes were dominant in both the FI and FT groups. The concentrations of propanoic, valeric and hexanoic acids were lower in the FI group at 2 weeks, and the levels of isobutyric and valeric acids were lower at 4 weeks after birth (P<0.05). The areas under the curves (AUCs) of propanoic, butanoic and valeric acids in predicting FI were 0.878, 0.816 and 0.744, respectively. Compared with that in the FT group, the relative bioluminescence of AI-2 was lower in the FI group at 2 weeks (P<0.05), and the AUC was 0.736. Conclusions The main composition of the microbiota was not obviously different in infants with FI. Some SCFAs and AI-2 have moderate value in predicting FI.
Collapse
Affiliation(s)
- Xiao-Chen Liu
- Neonatal Diagnosis and Treatment Centre of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Qian Sun
- Neonatal Diagnosis and Treatment Centre of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Yan-Chun Ji
- Neonatal Diagnosis and Treatment Centre of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Li-Zhen Fu
- Neonatal Diagnosis and Treatment Centre of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Zheng-Li Wang
- Neonatal Diagnosis and Treatment Centre of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Yu He
- Neonatal Diagnosis and Treatment Centre of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Lu-Quan Li
- Neonatal Diagnosis and Treatment Centre of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
- *Correspondence: Lu-Quan Li,
| |
Collapse
|
45
|
Bozzi Cionci N, Lucaccioni L, Pietrella E, Ficara M, Spada C, Torelli P, Bedetti L, Lugli L, Di Gioia D, Berardi A. Antibiotic Exposure, Common Morbidities and Main Intestinal Microbial Groups in Very Preterm Neonates: A Pilot Study. Antibiotics (Basel) 2022; 11:237. [PMID: 35203839 PMCID: PMC8868158 DOI: 10.3390/antibiotics11020237] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 01/27/2023] Open
Abstract
Prematurity exposes newborns to increased risks of infections and it is associated with critical morbidities. Preterm infants often require antibiotic therapies that can affect the correct establishment of gut microbiota. The aim of this study was to investigate targeted intestinal bacteria in preterm neonates with common morbidities and receiving antibiotic treatments of variable duration. Stool samples were collected after birth, at 15, 30 and 90 days of life. qPCR quantification of selected microbial groups (Bifidobacterium spp., Bacteroides fragilis group, Enterobacteriaceae, Clostridium cluster I and total bacteria) was performed and correlation between their levels, the duration of antibiotic treatment and different clinical conditions was studied. An increasing trend over time was observed for all microbial groups, especially for Bifdobacterium spp. Prolonged exposure to antibiotics in the first weeks of life affected Clostridium and B. fragilis levels, but these changes no longer persisted at 90 days of life. Variations of bacterial counts were associated with the length of hospital stay, feeding and mechanical ventilation. Late-onset sepsis and patent ductus arteriosus reduced the counts of Bifidobacterium, whereas B. fragilis was influenced by compromised respiratory conditions. This study can be a start point for the identification of microbial biomarkers associated with some common morbidities and tailored strategies for a healthy microbial development.
Collapse
Affiliation(s)
- Nicole Bozzi Cionci
- Department of Agricultural and Food Sciences, University of Bologna, 40127 Bologna, Italy; (N.B.C.); (D.D.G.)
| | - Laura Lucaccioni
- Pediatric Unit, Department of Medical and Surgical Sciences for Mothers, Children and Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Elisa Pietrella
- Pediatric Unit, Department of Medical for Mothers and Children, Ramazzini Hospital, 41012 Carpi, Italy;
| | - Monica Ficara
- Pediatric Unit, Department of Medical for Mothers and Children, Bufalini Hospital, 47521 Cesena, Italy;
| | - Caterina Spada
- Neonatal Intensive Care Unit, Department of Medical for Mothers and Children, Bufalini Hospital, 47521 Cesena, Italy;
| | - Paola Torelli
- Neonatal Intensive Care Unit, Department of Medical and Surgical Sciences for Mothers, Children and Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy; (P.T.); (L.B.); (L.L.); (A.B.)
| | - Luca Bedetti
- Neonatal Intensive Care Unit, Department of Medical and Surgical Sciences for Mothers, Children and Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy; (P.T.); (L.B.); (L.L.); (A.B.)
- PhD Program in Clinical and Experimental Medicine, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Licia Lugli
- Neonatal Intensive Care Unit, Department of Medical and Surgical Sciences for Mothers, Children and Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy; (P.T.); (L.B.); (L.L.); (A.B.)
| | - Diana Di Gioia
- Department of Agricultural and Food Sciences, University of Bologna, 40127 Bologna, Italy; (N.B.C.); (D.D.G.)
| | - Alberto Berardi
- Neonatal Intensive Care Unit, Department of Medical and Surgical Sciences for Mothers, Children and Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy; (P.T.); (L.B.); (L.L.); (A.B.)
| |
Collapse
|
46
|
Lu Y, Cai X, Zheng Y, Lyv Q, Wu J. Dominant bacteria and influencing factors of early intestinal colonization in very low birth weight infants: A prospective cohort study. J Clin Lab Anal 2022; 36:e24290. [PMID: 35148012 PMCID: PMC8906041 DOI: 10.1002/jcla.24290] [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: 01/04/2022] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 12/01/2022] Open
Abstract
Background The intestine of newborns is colonized by bacteria immediately after birth. This study explored dominant bacteria and influencing factors of early intestinal colonization in the early life of very low birth weight infants (VLBWI). Methods We enrolled 81 VLBWI and collected anal swabs at 24 h, 7th, 14th and 21st day after birth. We conducted bacterial culture for anal swabs, then selected the colony with obvious growth advantages in the plate for further culture and identification. Afterward, we analyzed the distribution and influencing factors of intestinal dominant microbiota combined with clinical data. Results A total of 300 specimens were collected, of which 62.67% (188/300) had obvious dominant bacteria, including 29.26% (55/188) Gram‐positive bacteria and 70.74% (133/188) Gram‐negative bacteria. The top five bacteria with the highest detection rates were Klebsiella pneumoniae, Escherichia coli, Enterococcus faecium, Enterococcus faecalis and Serratia marcescens. Meconium‐stained amniotic fluid and chorioamnionitis were correlated with intestinal bacterial colonization within 24 h of birth. Mechanical ventilation and antibiotics were independent risk factors affecting colonization. Nosocomial infection of K. pneumoniae and S. marcescens were associated with intestinal colonization. The colonization rates of K. pneumoniae, E. coli, E. faecium, and E. faecalis increased with the birth time. Conclusions The colonization rate in the early life of VLBWI increased over time and the predominant bacteria were Gram‐negative bacteria. Meconium‐stained amniotic fluid and chorioamnionitis affect intestinal colonization in early life. Mechanical ventilation and antibiotics were independent risk factors for intestinal bacterial colonization. The nosocomial infection of some bacteria was significantly related to intestinal colonization.
Collapse
Affiliation(s)
- Yanbo Lu
- School of Medicine, Ningbo University, Ningbo, China
| | - Xiaohong Cai
- School of Medicine, Ningbo University, Ningbo, China
| | - Yao Zheng
- Ningbo Women and Children's Hospital, Ningbo, China
| | - Qin Lyv
- Ningbo Women and Children's Hospital, Ningbo, China
| | - Junhua Wu
- Ningbo Women and Children's Hospital, Ningbo, China
| |
Collapse
|
47
|
Shah PA, Govindarajan V, Diggikar S, Rangaiah A, Devadas S, Kariyappa M. Exploring the Skin Mycobiome in Very Preterm babies during the early neonatal period in a Neonatal Intensive Care Unit of India. Trop Doct 2022; 52:362-364. [PMID: 35125017 DOI: 10.1177/00494755221077520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The neonatal skin microbiome consists of all the genomes and genetic products of micro-organisms harbouring the skin of babies. Host and the microbiota develop a harmonious environment resulting in symbiosis. Any disruption of this environment could lead to pathological disease. Our study was conducted to explore the neonatal skin fungal microbiome of very preterm neonates admitted to Neonatal Intensive Care Unit at a tertiary health care setting using Next Generation Sequencing of the18S rRNA gene. The most abundant genera found in 22/30 samples were Candida followed by Bipolaris & Cladosporium on the skin microbiome of these neonates. The presence of these fungi, whether just as commensals or as potential pathogens, is currently under research, owing to the risk of early exposure and incidence of infection right from birth.
Collapse
Affiliation(s)
- Pritik A Shah
- Department of Microbiology, Victoria Hospital, 29099Bangalore Medical College and Research Institute, Bengaluru, India.,Department of Paediatrics, Vani Vilas Hospital, 29099Bangalore Medical College and Research Institute, Bengaluru, India.,Pritik A Shah, Varun Govindarajan contributed equally
| | - Varun Govindarajan
- Department of Paediatrics, Vani Vilas Hospital, 29099Bangalore Medical College and Research Institute, Bengaluru, India.,Pritik A Shah, Varun Govindarajan contributed equally
| | - Shivshankar Diggikar
- Department of Neonatology, Ovum Woman and Child Specialty Hospital, Bengaluru, India
| | - Ambica Rangaiah
- Department of Microbiology, Victoria Hospital, 29099Bangalore Medical College and Research Institute, Bengaluru, India
| | - Sahana Devadas
- Department of Paediatrics, Vani Vilas Hospital, 29099Bangalore Medical College and Research Institute, Bengaluru, India
| | - Mallesh Kariyappa
- Department of Paediatrics, Vani Vilas Hospital, 29099Bangalore Medical College and Research Institute, Bengaluru, India
| |
Collapse
|
48
|
Obiero CW, Gumbi W, Mwakio S, Mwangudzah H, Seale AC, Taniuchi M, Liu J, Houpt E, Berkley JA. Detection of pathogens associated with early-onset neonatal sepsis in cord blood at birth using quantitative PCR. Wellcome Open Res 2022; 7:3. [PMID: 35600002 PMCID: PMC9114825 DOI: 10.12688/wellcomeopenres.17386.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2021] [Indexed: 12/20/2022] Open
Abstract
Background: Early onset neonatal sepsis (EONS) typically begins prior to, during or soon after birth and may be rapidly fatal. There is paucity of data on the aetiology of EONS in sub-Saharan Africa due to limited diagnostic capacity in this region, despite the associated significant mortality and long-term neurological impairment. Methods: We compared pathogens detected in cord blood samples between neonates admitted to hospital with possible serious bacterial infection (pSBI) in the first 48 hours of life (cases) and neonates remaining well (controls). Cord blood was systematically collected at Kilifi County Hospital (KCH) from 2011-2016, and later tested for 21 bacterial, viral and protozoal targets using multiplex PCR via TaqMan Array Cards (TAC). Results: Among 603 cases (101 [17%] of whom died), 179 (30%) tested positive for ≥1 target and 37 (6.1%) tested positive for multiple targets. Klebsiella oxytoca, Escherichia coli/Shigella spp., Pseudomonas aeruginosa, and Streptococcus pyogenes were commonest. Among 300 controls, 79 (26%) tested positive for ≥1 target, 11 (3.7%) were positive for multiple targets, and K. oxytoca and P. aeruginosa were most common. Cumulative odds ratios across controls: cases (survived): cases (died) were E. coli/Shigella spp. 2.6 (95%CI 1.6-4.4); E. faecalis 4.0 (95%CI 1.1-15); S. agalactiae 4.5 (95%CI 1.6-13); Ureaplasma spp. 2.9 (95%CI 1.3-6.4); Enterovirus 9.1 (95%CI 2.3-37); and Plasmodium spp. 2.9 (95%CI 1.4-6.2). Excluding K. oxytoca and P. aeruginosa as likely contaminants, aetiology was attributed in 9.4% (95%CI 5.1-13) cases using TAC. Leading pathogen attributions by TAC were E. coli/Shigella spp. (3.5% (95%CI 1.7-5.3)) and Ureaplasma spp. (1.7% (95%CI 0.5-3.0)). Conclusions: Cord blood sample may be useful in describing EONS pathogens at birth, but more specific tests are needed for individual diagnosis. Careful sampling of cord blood using aseptic techniques is crucial to minimize contamination. In addition to culturable bacteria, Ureaplasma and Enterovirus were causes of EONS.
Collapse
Affiliation(s)
- Christina W. Obiero
- Clinical research, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Global health, Academic Medical Center of the University of Amsterdam, Amsterdam, The Netherlands
| | - Wilson Gumbi
- Bioscience department, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Stella Mwakio
- Clinical research, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Hope Mwangudzah
- Clinical research, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Anna C. Seale
- Clinical research, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
- College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Mami Taniuchi
- Division of Infectious Diseases and International Health, University of Virginia, Virginia, USA
| | - Jie Liu
- Division of Infectious Diseases and International Health, University of Virginia, Virginia, USA
| | - Eric Houpt
- Division of Infectious Diseases and International Health, University of Virginia, Virginia, USA
| | - James A. Berkley
- Clinical research, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine, University of Oxford, Oxford, UK
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi, Kenya
| |
Collapse
|
49
|
Babić S, Jovandarić M. The main features of placental abruption: Clinical presentation and treatment. SRPSKI MEDICINSKI CASOPIS LEKARSKE KOMORE 2022; 3:446-454. [DOI: 10.5937/smclk3-37202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Placental abruption is defined as the premature separation of the placenta from the uterus. Rapid diagnosis and adequate medical intervention provide a good outcome, which is not possible in all countries. Any bleeding that occurs in the second half of pregnancy raises the suspicion of placental abruption, which is associated with fetal and maternal morbidity and mortality. The clinical picture and consequences for the fetus depend on the degree of bleeding and the amount of blood. Complete abruption leads to the disruption of fetal circulation and oxygenation, asphyxia, and stillbirth. In case of partial abruption, the consequences for the fetus depend on the degree of bleeding. In complete abruption, fetal death is inevitable unless an emergency cesarean delivery is undertaken.
Collapse
|
50
|
Spontaneous preterm delivery is reflected in both early neonatal and maternal gut microbiota. Pediatr Res 2022; 91:1804-1811. [PMID: 34349229 PMCID: PMC9270225 DOI: 10.1038/s41390-021-01663-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 04/22/2021] [Accepted: 07/06/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Aberrant gut microbiota composition in preterm neonates is linked to adverse health consequences. Little is known about the impact of perinatal factors or maternal gut microbiota on initial preterm gut colonization. METHODS Fecal samples were collected from 55 preterm neonates (<35 gestational weeks), 51 mothers, and 25 full-term neonates during the first 3-4 postpartum days. Gut microbiota composition was assessed using 16S ribosomal RNA gene sequencing. RESULTS Preterm neonates exhibited significantly lower gut microbiota alpha diversity and distinct beta diversity clustering compared to term neonates. Spontaneous preterm birth was associated with distinct initial gut microbiota beta diversity as compared to iatrogenic delivery. Gestational age or delivery mode had no impact on the preterm gut microbiota composition. The cause of preterm delivery was also reflected in the maternal gut microbiota composition. The contribution of maternal gut microbiota to initial preterm gut colonization was more pronounced after spontaneous delivery than iatrogenic delivery and not dependent on delivery mode. CONCLUSIONS The initial preterm gut microbiota is distinct from term microbiota. Spontaneous preterm birth is reflected in the early neonatal and maternal gut microbiota. Transmission of gut microbes from mother to neonate is determined by spontaneous preterm delivery, but not by mode of birth. IMPACT The initial gut microbiota in preterm neonates is distinct from those born full term. Spontaneous preterm birth is associated with changes in the gut microbiota composition of both preterm neonates and their mothers. The contribution of the maternal gut microbiota to initial neonatal gut colonization was more pronounced after spontaneous preterm delivery as compared to iatrogenic preterm delivery and not dependent on delivery mode. Our study provides new evidence regarding the early gut colonization patterns in preterm infants. Altered preterm gut microbiota has been linked to adverse health consequences and may provide a target for early intervention.
Collapse
|