1
|
Sha C, Jin Z, Ku SY, Kogosov AS, Yu S, Bergese SD, Hsieh H. Necrotizing Enterocolitis and Neurodevelopmental Impairments: Microbiome, Gut, and Brain Entanglements. Biomolecules 2024; 14:1254. [PMID: 39456187 PMCID: PMC11505939 DOI: 10.3390/biom14101254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024] Open
Abstract
There is significant communication and interdependence among the gut, the microbiome, and the brain during development. Diseases, such as necrotizing enterocolitis (NEC), highlight how injury to the immature gastrointestinal tract leads to long-term neurological consequences, due to vulnerabilities of the brain in the early stages of life. A better understanding of the developing gut-microbiota-brain axis is needed to both prevent and treat the devastating consequences of these disease processes. The gut-microbiota-brain axis is a bidirectional communication pathway that includes metabolic, nervous, endocrine, and immune components. In this review, we discuss gut development, microbiome colonization and maturation, and the interactions that influence neurodevelopment in the context of NEC. We describe the components of the gut-brain axis and how the microbiome is an integral member of this relationship. Finally, we explore how derangements within the microbiome and gut-microbiota-brain axis affect the normal development and function of the other systems and long-term neurodevelopmental consequences for patients.
Collapse
Affiliation(s)
- Cuilee Sha
- Department of Pharmacological Sciences, Stony Brook University, 100 Nicolls Road, Stony Brook, NY 11794, USA;
- Center for Nervous System Disorders, Stony Brook University, 100 Nicolls Road, Stony Brook, NY 11794, USA
| | - Zhaosheng Jin
- Department of Anesthesiology, Stony Brook Medicine, 101 Nicolls Road, Stony Brook, NY 11794, USA;
| | - Stella Y. Ku
- Stony Brook University, 100 Nicolls Road, Stony Brook, NY 11794, USA
| | - Ann S. Kogosov
- Renaissance School of Medicine, Stony Brook University, 100 Nicolls Road, Stony Brook, NY 11794, USA
| | - Sun Yu
- Department of Surgery, Stony Brook Medicine, 101 Nicolls Road, Stony Brook, NY 11794, USA
| | - Sergio D. Bergese
- Department of Anesthesiology, Stony Brook Medicine, 101 Nicolls Road, Stony Brook, NY 11794, USA;
| | - Helen Hsieh
- Center for Nervous System Disorders, Stony Brook University, 100 Nicolls Road, Stony Brook, NY 11794, USA
- Department of Surgery, Stony Brook Medicine, 101 Nicolls Road, Stony Brook, NY 11794, USA
| |
Collapse
|
2
|
Malamitsi-Puchner A, Briana DD, Di Renzo GC. The microbiome in pregnancy and early life-Highlights from the 11th Maria Delivoria-Papadopoulos Perinatal Symposium. Acta Paediatr 2024; 113:2189-2196. [PMID: 38895845 DOI: 10.1111/apa.17328] [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: 04/02/2024] [Revised: 06/05/2024] [Accepted: 06/11/2024] [Indexed: 06/21/2024]
Abstract
This review was based on a symposium that examined novel aspects of the microbiome during pregnancy and early life and explored papers published by the lecturers. For example, it showed that bacterial extracellular vesicles derived from the microbiome harboured in various maternal niches, carried bacterial deoxyribonucleic acid, were isolated from the placenta and may have confounded placental microbiome studies. Maternal diet was responsible for the composition and diversity of breast milk microbiota, and may have shaped the offspring's microbiome and influenced their immune components. Probiotics and antibiotics administered perinatally may have had beneficial but also long-lasting adverse effects on offspring.
Collapse
Affiliation(s)
- Ariadne Malamitsi-Puchner
- Neonatal Intensive Care Unit, 3rd Department of Pediatrics, National and Kapodistrian University of Athens, Athens, Greece
| | - Despina D Briana
- Neonatal Intensive Care Unit, 3rd Department of Pediatrics, National and Kapodistrian University of Athens, Athens, Greece
| | - Gian Carlo Di Renzo
- PREIS School, International and European School of Perinatal, Neonatal and Reproductive Medicine, Florence, Italy
- Department of Obstetrics, Gynecology and Perinatology, I.M. Sechenov First State University of Moscow, Moscow, Russia
| |
Collapse
|
3
|
Dera N, Żeber-Lubecka N, Ciebiera M, Kosińska-Kaczyńska K, Szymusik I, Massalska D, Dera K, Bubień K. Intrauterine Shaping of Fetal Microbiota. J Clin Med 2024; 13:5331. [PMID: 39274545 PMCID: PMC11396688 DOI: 10.3390/jcm13175331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 08/18/2024] [Accepted: 08/29/2024] [Indexed: 09/16/2024] Open
Abstract
Mechanisms resulting from the physiological immaturity of the digestive system in children delivered before 32 weeks of gestation and, in particular, different interactions between the microbiome and the body have not been fully elucidated yet. Next-generation sequencing methods demonstrated the presence of bacterial DNA in the placenta and amniotic fluid, which may reflect bacterial populations that initiate intestinal colonization in utero. Numerous studies confirmed the hypothesis stating that intestinal bacteria played an important role in the pathogenesis of necrotizing enterocolitis (NEC) early- and late-onset neonatal sepsis (EONS and LONS). The model and scale of disorders within the intestinal microbiome are the subject of active research in premature infants. Neonatal meconium was primarily used as an indicator defining the environment in utero, as it is formed before birth. Metagenomic results and previous data from microbiological bacterial cultures showed a correlation between the time from birth to sample collection and the detection of bacteria in the neonatal meconium. Therefore, it may be determined that the colonization of the newborn's intestines is influenced by numerous factors, which may be divided into prenatal, perinatal, and postnatal, with particular emphasis put on the mode of delivery and contact with the parent immediately after birth. Background: The aim of this review was to collect available data on the intrauterine shaping of the fetal microbiota. Methods: On 13 March 2024, the available literature in the PubMed National Library of Medicine search engine was reviewed using the following selected keywords: "placental microbiome", "intestinal bacteria in newborns and premature infants", and "intrauterine microbiota". Results: After reviewing the available articles and abstracts and an in-depth analysis of their content, over 100 articles were selected for detailed elaboration. We focused on the origin of microorganisms shaping the microbiota of newborns. We also described the types of bacteria that made up the intrauterine microbiota and the intestinal microbiota of newborns. Conclusions: The data presented in the review on the microbiome of both term newborns and those with a body weight below 1200 g indicate a possible intrauterine colonization of the fetus depending on the duration of pregnancy. The colonization occurs both via the vaginal and intestinal route (hematogenous route). However, there are differences in the demonstrated representatives of various types of bacteria, phyla Firmicutes and Actinobacteria in particular, taking account of the distribution in their abundance in the individual groups of pregnancy duration. Simultaneously, the distribution of the phyla Actinobacteria and Proteobacteria is consistent. Considering the duration of pregnancy, it may also be concluded that the bacterial flora of vaginal origin dominates in preterm newborns, while the flora of intestinal origin dominates in term newborns. This might explain the role of bacterial and infectious factors in inducing premature birth with the rupture of fetal membranes.
Collapse
Affiliation(s)
- Norbert Dera
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, 01-809 Warsaw, Poland
- Warsaw Institute of Women's Health, 00-189 Warsaw, Poland
| | - Natalia Żeber-Lubecka
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, 02-781 Warsaw, Poland
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Michał Ciebiera
- Warsaw Institute of Women's Health, 00-189 Warsaw, Poland
- Second Department of Obstetrics and Gynecology, Centre of Postgraduate Medical Education, 00-189 Warsaw, Poland
| | - Katarzyna Kosińska-Kaczyńska
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, 01-809 Warsaw, Poland
| | - Iwona Szymusik
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, 01-809 Warsaw, Poland
| | - Diana Massalska
- Warsaw Institute of Women's Health, 00-189 Warsaw, Poland
- Second Department of Obstetrics and Gynecology, Centre of Postgraduate Medical Education, 00-189 Warsaw, Poland
| | - Kacper Dera
- Provincial Specialist Hospital in Olsztyn, 10-561 Olsztyn, Poland
| | - Katarzyna Bubień
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, 01-809 Warsaw, Poland
| |
Collapse
|
4
|
Lai MY, Chang YH, Lee CC. The impact of gut microbiota on morbidities in preterm infants. Kaohsiung J Med Sci 2024; 40:780-788. [PMID: 39073226 DOI: 10.1002/kjm2.12878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024] Open
Abstract
The gut microbiota undergoes substantial development from birth, and its development in the initial years of life has a potentially lifelong effect on the health of the individual. However, various factors can disrupt the development of the gut microbiota, leading to a condition known as dysbiosis, particularly in preterm infants. Current studies involving adults have suggested that the gut microbiota not only influences the gut but also has multidimensional effects on remote organs; these pathways are often referred to as the gut-organ axis. Imbalance of the gut microbiota may lead to the development of multiple diseases. Recent studies have revealed that gut dysbiosis in preterm infants may cause several acute morbidities-such as necrotizing enterocolitis, late-onset sepsis, bronchopulmonary dysplasia, and retinopathy of prematurity-and it may also influence long-term outcomes including neurodevelopment and somatic growth. This review mainly presents the existing evidence regarding the relationships between the gut microbiota and these morbidities in preterm infants and explores the role of the gut-organ axis in these morbidities. This paper thus offers insights into the future perspectives on microbiota interventions for promoting the health of preterm infants.
Collapse
Affiliation(s)
- Mei-Yin Lai
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital, School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yin-Hsi Chang
- Department of Ophthalmology, Chang Gung Memorial Hospital, School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chien-Chung Lee
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital, School of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
5
|
Veseli I, Chen YT, Schechter MS, Vanni C, Fogarty EC, Watson AR, Jabri BA, Blekhman R, Willis AD, Yu MK, Fernandez-Guerra A, Fussel J, Eren AM. Microbes with higher metabolic independence are enriched in human gut microbiomes under stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.10.540289. [PMID: 37293035 PMCID: PMC10245760 DOI: 10.1101/2023.05.10.540289] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
A wide variety of human diseases are associated with loss of microbial diversity in the human gut, inspiring a great interest in the diagnostic or therapeutic potential of the microbiota. However, the ecological forces that drive diversity reduction in disease states remain unclear, rendering it difficult to ascertain the role of the microbiota in disease emergence or severity. One hypothesis to explain this phenomenon is that microbial diversity is diminished as disease states select for microbial populations that are more fit to survive environmental stress caused by inflammation or other host factors. Here, we tested this hypothesis on a large scale, by developing a software framework to quantify the enrichment of microbial metabolisms in complex metagenomes as a function of microbial diversity. We applied this framework to over 400 gut metagenomes from individuals who are healthy or diagnosed with inflammatory bowel disease (IBD). We found that high metabolic independence (HMI) is a distinguishing characteristic of microbial communities associated with individuals diagnosed with IBD. A classifier we trained using the normalized copy numbers of 33 HMI-associated metabolic modules not only distinguished states of health versus IBD, but also tracked the recovery of the gut microbiome following antibiotic treatment, suggesting that HMI is a hallmark of microbial communities in stressed gut environments.
Collapse
|
6
|
Inchingolo F, Inchingolo AM, Latini G, Ferrante L, de Ruvo E, Campanelli M, Longo M, Palermo A, Inchingolo AD, Dipalma G. Difference in the Intestinal Microbiota between Breastfeed Infants and Infants Fed with Artificial Milk: A Systematic Review. Pathogens 2024; 13:533. [PMID: 39057760 PMCID: PMC11280328 DOI: 10.3390/pathogens13070533] [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: 05/29/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
The gut microbiota (GM) plays a crucial role in human health, particularly during the first years of life. Differences in GM between breastfed and formula (F)-fed infants may influence long-term health outcomes. This systematic review aims to compare the gut microbiota of breastfed infants with that of F-fed infants and to evaluate the clinical implications of these differences. We searched databases on Scopus, Web of Science, and Pubmed with the following keywords: "gut microbiota", "gut microbiome", and "neonatal milk". The inclusion criteria were articles relating to the analysis of the intestinal microbiome of newborns in relation to the type of nutrition, clinical studies or case series, excluding reviews, meta-analyses, animal models, and in vitro studies. The screening phase ended with the selection of 13 publications for this work. Breastfed infants showed higher levels of beneficial bacteria such as Bifidobacterium and Lactobacillus, while F-fed infants had a higher prevalence of potentially pathogenic bacteria, including Clostridium difficile and Enterobacteriaceae. Infant feeding type influences the composition of oral GM significantly. Breastfeeding promotes a healthier and more diverse microbial ecosystem, which may offer protective health benefits. Future research should explore strategies to improve the GM of F-fed infants and understand the long-term health implications.
Collapse
Affiliation(s)
- Francesco Inchingolo
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy or (A.M.I.); or (G.L.); or (L.F.); or (E.d.R.); (M.L.); or (A.D.I.); or (G.D.)
| | - Angelo Michele Inchingolo
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy or (A.M.I.); or (G.L.); or (L.F.); or (E.d.R.); (M.L.); or (A.D.I.); or (G.D.)
| | - Giulia Latini
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy or (A.M.I.); or (G.L.); or (L.F.); or (E.d.R.); (M.L.); or (A.D.I.); or (G.D.)
| | - Laura Ferrante
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy or (A.M.I.); or (G.L.); or (L.F.); or (E.d.R.); (M.L.); or (A.D.I.); or (G.D.)
| | - Elisabetta de Ruvo
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy or (A.M.I.); or (G.L.); or (L.F.); or (E.d.R.); (M.L.); or (A.D.I.); or (G.D.)
| | - Merigrazia Campanelli
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy or (A.M.I.); or (G.L.); or (L.F.); or (E.d.R.); (M.L.); or (A.D.I.); or (G.D.)
| | - Marialuisa Longo
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy or (A.M.I.); or (G.L.); or (L.F.); or (E.d.R.); (M.L.); or (A.D.I.); or (G.D.)
| | - Andrea Palermo
- College of Medicine and Dentistry, Birmingham B4 6BN, UK;
| | - Alessio Danilo Inchingolo
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy or (A.M.I.); or (G.L.); or (L.F.); or (E.d.R.); (M.L.); or (A.D.I.); or (G.D.)
| | - Gianna Dipalma
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy or (A.M.I.); or (G.L.); or (L.F.); or (E.d.R.); (M.L.); or (A.D.I.); or (G.D.)
| |
Collapse
|
7
|
Hick E, Suárez M, Rey A, Mantecón L, Fernández N, Solís G, Gueimonde M, Arboleya S. Personalized Nutrition with Banked Human Milk for Early Gut Microbiota Development: In Pursuit of the Perfect Match. Nutrients 2024; 16:1976. [PMID: 38999725 PMCID: PMC11243202 DOI: 10.3390/nu16131976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/17/2024] [Accepted: 06/19/2024] [Indexed: 07/14/2024] Open
Abstract
The correct initial colonization and establishment of the gut microbiota during the early stages of life is a key step, with long-lasting consequences throughout the entire lifespan of the individual. This process is affected by several perinatal factors; among them, feeding mode is known to have a critical role. Breastfeeding is the optimal nutrition for neonates; however, it is not always possible, especially in cases of prematurity or early pathology. In such cases, most commonly babies are fed with infant formulas in spite of the official nutritional and health international organizations' recommendation on the use of donated human milk through milk banks for these cases. However, donated human milk still does not totally match maternal milk in terms of infant growth and gut microbiota development. The present review summarizes the practices of milk banks and hospitals regarding donated human milk, its safety and quality, and the health outcomes in infants fed with donated human milk. Additionally, we explore different alternatives to customize pasteurized donated human milk with the aim of finding the perfect match between each baby and banked milk for promoting the establishment of a beneficial gut microbiota from the early stages of life.
Collapse
Affiliation(s)
- Emilia Hick
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), 33300 Villaviciosa, Spain
| | - Marta Suárez
- Pediatrics Service, Central University Hospital of Asturias (HUCA-SESPA), 33011 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Alejandra Rey
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), 33300 Villaviciosa, Spain
| | - Laura Mantecón
- Pediatrics Service, Central University Hospital of Asturias (HUCA-SESPA), 33011 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Nuria Fernández
- Pediatrics Service, University Hospital of Cabueñes (CAB-SESPA), 33394 Gijón, Spain
| | - Gonzalo Solís
- Pediatrics Service, Central University Hospital of Asturias (HUCA-SESPA), 33011 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), 33300 Villaviciosa, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Silvia Arboleya
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), 33300 Villaviciosa, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| |
Collapse
|
8
|
Moreno-Sanz B, Lázaro-Perona F, Escribano E, Cabrera Lafuente M, Montes MT, Amorós R, Arboleya S, Gueimonde M, Mingorance J, Saenz de Pipaon M. Assessment trial of the effect of enteral insulin on the preterm infant intestinal microbiota. Pediatr Res 2024; 95:1117-1123. [PMID: 38086952 DOI: 10.1038/s41390-023-02917-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 10/05/2023] [Accepted: 10/25/2023] [Indexed: 03/09/2024]
Abstract
BACKGROUND Insulin might be associated with changes in infant gastrointestinal microbiota. The objective of this randomized controlled trial was to assess the efficacy of two doses of recombinant human(rh) enteral insulin administration compared to placebo in intestinal microbiota. METHODS 19 preterm patients were recruited at the NICU of La Paz University Hospital (Madrid, Spain). Subjects received 2000 µIU of rh enteral insulin/ml(n = 8), 400 µIU of rh enteral insulin/ml(n = 6) or placebo(n = 5) for 28 days administered once per day. Extracted DNA from fecal samples collected at the beginning and end of treatment were analyzed. The 16S rRNA V4 region was amplified and sequenced in a Miseq(Illumina®) sequencer using 2 × 250 bp paired end. Resulting reads were filtered and analyzed using Qiime2 software. Metabolic activity was assessed by GC. RESULTS Gestational age and birth weight did not differ between groups. At the phylum level, both insulin treated groups increased the relative abundance of Bacillota, while Pseudomonadota decreased. No change was observed in infants receiving placebo. At the genus level, insulin at both doses showed enriching effects on Clostridium. We found a significant increase in concentrations of fecal propionate in both rh insulin treated groups. CONCLUSION Rh insulin may modify neonatal intestinal microbiota and SCFAs in preterm infants. IMPACT STATEMENT Decrease of Pseudomonadota (former Proteobacteria phylum) and increase of Bacillota (former Firmicutes phylum) obtained in this study are the changes observed previously in low-risk infants for NEC. The administration of recombinant enteral insulin may modify the microbiota of preterm new-borns and SCFAs. Modulation of the microbiota may be a mechanism whereby insulin contributes to neonatal intestinal maturation and/or protection.
Collapse
Affiliation(s)
- Bárbara Moreno-Sanz
- Department of Neonatology, La Paz University Hospital Health Research Institute, Madrid, Spain
| | - Fernando Lázaro-Perona
- Department of Microbiology, La Paz University Hospital Health Research Institute, Madrid, Spain
| | - Esperanza Escribano
- Department of Neonatology, La Paz University Hospital Health Research Institute, Madrid, Spain
| | - Marta Cabrera Lafuente
- Department of Neonatology, La Paz University Hospital Health Research Institute, Madrid, Spain
| | - María Teresa Montes
- Department of Neonatology, La Paz University Hospital Health Research Institute, Madrid, Spain
| | - Rocío Amorós
- Department of Neonatology, La Paz University Hospital Health Research Institute, Madrid, Spain
| | - Silvia Arboleya
- Department of Microbiology and Biochemistry of Dairy Products, IPLA-CSIC, Villaviciosa, Spain
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry of Dairy Products, IPLA-CSIC, Villaviciosa, Spain
| | - Jesús Mingorance
- Department of Microbiology, La Paz University Hospital Health Research Institute, Madrid, Spain
| | - Miguel Saenz de Pipaon
- Department of Neonatology, La Paz University Hospital Health Research Institute, Madrid, Spain.
- Universidad Autonoma de Madrid Madrid, Madrid, Spain.
| |
Collapse
|
9
|
Cifuentes MP, Chapman JA, Stewart CJ. Gut microbiome derived short chain fatty acids: Promising strategies in necrotising enterocolitis. CURRENT RESEARCH IN MICROBIAL SCIENCES 2024; 6:100219. [PMID: 38303965 PMCID: PMC10831176 DOI: 10.1016/j.crmicr.2024.100219] [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] [Indexed: 02/03/2024] Open
Abstract
Necrotising enterocolitis (NEC) is a devastating condition that poses a significant risk of morbidity and mortality, particularly among preterm babies. Extensive research efforts have been directed at identifying optimal treatment and diagnostic strategies but results from such studies remain unclear and controversial. Among the most promising candidates are prebiotics, probiotics and their metabolites, including short chain fatty acids (SCFAs). Such metabolites have been widely explored as possible biomarkers of gut health for different clinical conditions, with overall positive effects on the host observed. This review aims to describe the role of gut microbiome derived SCFAs in necrotising enterocolitis. Until now, information has been conflicting, with the primary focus on the main three SCFAs (acetic acid, propionic acid, and butyric acid). While numerous studies have indicated the relationship between SCFAs and NEC, the current evidence is insufficient to draw definitive conclusions about the use of these metabolites as NEC biomarkers or their potential in treatment strategies. Ongoing research in this area will help enhance both our understanding of SCFAs as valuable indicators of NEC and their practical application in clinical settings.
Collapse
Affiliation(s)
- María P Cifuentes
- Translational and Clinical Research Institute, Newcastle University, Newcastle. UK
| | - Jonathan A Chapman
- Translational and Clinical Research Institute, Newcastle University, Newcastle. UK
| | | |
Collapse
|
10
|
Hong L, Huang Y, Han J, Li S, Zhang L, Zhou Q, Cao X, Yu W, Guo X, Yang Y, Zhou Y, Yan W, Hong S, Jiang S, Cao Y. Pathogen-specific alterations in intestinal microbiota precede urinary tract infections in preterm infants: a longitudinal case-control study. Gut Microbes 2024; 16:2333413. [PMID: 38561312 PMCID: PMC10986765 DOI: 10.1080/19490976.2024.2333413] [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: 11/27/2023] [Accepted: 03/18/2024] [Indexed: 04/04/2024] Open
Abstract
Urinary tract infections (UTIs) are among the most common late-onset infections in preterm infants, characterized by nonspecific symptoms and a pathogenic spectrum that diverges from that of term infants and older children, which present unique diagnostic and therapeutic challenges. Existing data on the role of gut microbiota in UTI pathogenesis in this demographic are limited. This study aims to investigate alterations in gut microbiota and fecal calprotectin levels and their association with the development of UTIs in hospitalized preterm infants. A longitudinal case-control study was conducted involving preterm infants admitted between January 2018 and October 2020. Fecal samples were collected weekly and analyzed for microbial profiles and calprotectin levels. Propensity score matching, accounting for key perinatal factors including age and antibiotic use, was utilized to match samples from UTI-diagnosed infants to those from non-UTI counterparts. Among the 151 preterm infants studied, 53 were diagnosed with a UTI, predominantly caused by Enterobacteriaceae (79.3%) and Enterococcaceae (19.0%). Infants with UTIs showed a significantly higher abundance of these families compared to non-UTI infants, for both Gram-negative and positive pathogens, respectively. Notably, there was a significant pre-UTI increase in the abundance of pathogen-specific taxa in infants later diagnosed with UTIs, offering high predictive value for early detection. Shotgun metagenomic sequencing further confirmed the dominance of specific pathogenic species pre-UTI and revealed altered virulence factor profiles associated with Klebsiella aerogenes and Escherichia coli infections. Additionally, a decline in fecal calprotectin levels was observed preceding UTI onset, particularly in cases involving Enterobacteriaceae. The observed pathogen-specific alterations in the gut microbiota preceding UTI onset offer novel insight into the UTI pathogenesis and promising early biomarkers for UTIs in preterm infants, potentially enhancing the timely management of this common infection. However, further validation in larger cohorts is essential to confirm these findings.
Collapse
Affiliation(s)
- Luyang Hong
- Department of Neonatology, Children’s Hospital of Fudan University, Shanghai, China
| | - Yihuang Huang
- Department of Neonatology, Children’s Hospital of Fudan University, Shanghai, China
| | - Junyan Han
- Department of Neonatology, Children’s Hospital of Fudan University, Shanghai, China
| | - Shujuan Li
- Department of Neonatology, Children’s Hospital of Fudan University, Shanghai, China
| | - Lan Zhang
- Department of Neonatology, Children’s Hospital of Fudan University, Shanghai, China
| | - Qi Zhou
- Department of Neonatology, Children’s Hospital of Fudan University, Shanghai, China
| | - Xincheng Cao
- Department of Neonatology, Children’s Hospital of Fudan University, Shanghai, China
| | - Weiyin Yu
- Department of Neonatology, Children’s Hospital of Fudan University, Shanghai, China
| | - Xinhui Guo
- Department of Neonatology, Children’s Hospital of Fudan University, Shanghai, China
| | - Yi Yang
- NHC Key Laboratory of Neonatal Diseases, Children’s Hospital of Fudan University, Shanghai, China
| | - Yufeng Zhou
- NHC Key Laboratory of Neonatal Diseases, Children’s Hospital of Fudan University, Shanghai, China
| | - Weili Yan
- National Children’s Medical Center, Department of Clinical Epidemiology of Children’s Hospital of Fudan University, Shanghai, China
| | - Shangyu Hong
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Siyuan Jiang
- Department of Neonatology, Children’s Hospital of Fudan University, Shanghai, China
| | - Yun Cao
- Department of Neonatology, Children’s Hospital of Fudan University, Shanghai, China
- NHC Key Laboratory of Neonatal Diseases, Children’s Hospital of Fudan University, Shanghai, China
| |
Collapse
|
11
|
Bosco A, Piu C, Picciau ME, Pintus R, Fanos V, Dessì A. Metabolomics in NEC: An Updated Review. Metabolites 2023; 14:14. [PMID: 38248817 PMCID: PMC10821135 DOI: 10.3390/metabo14010014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 12/13/2023] [Accepted: 12/20/2023] [Indexed: 01/23/2024] Open
Abstract
Necrotizing enterocolitis (NEC) represents the most common and lethal acute gastrointestinal emergency of newborns, mainly affecting those born prematurely. It can lead to severe long-term sequelae and the mortality rate is approximately 25%. Furthermore, the diagnosis is difficult, especially in the early stages, due to multifactorial pathogenesis and complex clinical pictures with mild and non-specific symptoms. In addition, the existing tests have poor diagnostic value. Thus, the scientific community has been focusing its attention on the identification of non-invasive biomarkers capable of prediction, early diagnosis and discriminating NEC from other intestinal diseases in order to intervene early and block the progression of the pathology. In this regard, the use of "omics" technologies, especially metabolomics and microbiomics, could be a fundamental synergistic strategy to study the pathophysiology of NEC. In addition, a deeper knowledge of the microbiota-host cross-talk can clarify the metabolic pathways potentially involved in the pathology, allowing for the identification of specific biomarkers. In this article, the authors analyze the state-of-the-art concerning the application of metabolomics and microbiota analysis to investigate this pathology and discuss the future possibility of the metabolomic fingerprint of patients for diagnostic purposes.
Collapse
Affiliation(s)
| | | | | | | | | | - Angelica Dessì
- Department of Surgical Sciences, University of Cagliari and Neonatal Intensive Care Unit, AOU Cagliari, 09124 Cagliari, Italy; (A.B.); (C.P.); (M.E.P.); (R.P.); (V.F.)
| |
Collapse
|
12
|
Hong L, Huang Y, Han J, Li S, Zhang L, Jiang S, Zhou Q, Cao X, Yu W, Yang Y, Hong S, Zhou Y, Yan W, Cao Y. Dynamics and Crosstalk between Gut Microbiota, Metabolome, and Fecal Calprotectin in Very Preterm Infants: Insights into Feeding Intolerance. Nutrients 2023; 15:4849. [PMID: 38004243 PMCID: PMC10674500 DOI: 10.3390/nu15224849] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND Feeding intolerance (FI) is a significant concern in the care of preterm infants, impacting their growth and development. We previously reported that FI is linked to lower fecal calprotectin (FC) levels. This study aims to explore the postnatal dynamics and interplay between microbiota, metabolic profiles, and host immunity in preterm infants with and without FI. METHODS Infants with gestational age <32 weeks or birth weight <1500 g were enrolled at the Children's Hospital of Fudan University between January 2018 and October 2020. Weekly fecal samples were analyzed for bacterial profiling, metabolome, and calprotectin levels, exploring their longitudinal development and interrelationships. RESULTS Of the 118 very preterm infants studied, 48 showed FI. These infants experienced an interrupted microbial-immune trajectory, particularly at 3-4 weeks of age, marked by a reduced bacterial abundance, alpha diversity, and FC levels. Metabolic changes in FI were pronounced between 3 and 6 weeks. Pantothenic acid and two polyamine metabolites were closely associated with bacterial abundance and FC levels and negatively correlated with the duration to attain full enteral feeding. CONCLUSIONS FI infants demonstrated compromised microbiome-immune interactions, potentially influenced by specific metabolites. This research underscored the importance of early microbial and metabolic development in the pathogenesis of FI in very preterm infants.
Collapse
Affiliation(s)
- Luyang Hong
- Department of Neonatology, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai 201102, China
| | - Yihuang Huang
- Department of Neonatology, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai 201102, China
| | - Junyan Han
- Department of Neonatology, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai 201102, China
| | - Shujuan Li
- Department of Neonatology, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai 201102, China
| | - Lan Zhang
- Department of Neonatology, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai 201102, China
| | - Siyuan Jiang
- Department of Neonatology, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai 201102, China
| | - Qi Zhou
- Department of Neonatology, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai 201102, China
| | - Xincheng Cao
- Department of Neonatology, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai 201102, China
| | - Weiyin Yu
- Department of Neonatology, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai 201102, China
| | - Yi Yang
- NHC Key Laboratory of Neonatal Diseases, Fudan University, Shanghai 201102, China; (Y.Y.)
| | - Shangyu Hong
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China;
| | - Yufeng Zhou
- NHC Key Laboratory of Neonatal Diseases, Fudan University, Shanghai 201102, China; (Y.Y.)
| | - Weili Yan
- Department of Clinical Epidemiology, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai 201102, China
| | - Yun Cao
- Department of Neonatology, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai 201102, China
- NHC Key Laboratory of Neonatal Diseases, Fudan University, Shanghai 201102, China; (Y.Y.)
| |
Collapse
|
13
|
Zhang JY, Greenwald MJ, Rodriguez SH. Gut Microbiome and Retinopathy of Prematurity. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1683-1690. [PMID: 36780985 DOI: 10.1016/j.ajpath.2023.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/17/2023] [Accepted: 01/26/2023] [Indexed: 02/13/2023]
Abstract
Retinopathy of prematurity (ROP), a leading cause of childhood blindness worldwide, is strongly associated with gestational age and weight at birth. Yet, many extremely preterm infants never develop ROP or develop only mild ROP with spontaneous regression. In addition, a myriad of other factors play a role in the retinal pathology, one of which may include the early gut microbiome. The complications associated with early gestational age include dysbiosis of the dynamic neonatal gut microbiome, as evidenced by the development of often concomitant conditions, such as necrotizing enterocolitis. Given this, alongside growing evidence for a gut-retina axis, there is an increasing interest in how the early intestinal environment may play a role in the pathophysiology of ROP. Potential mechanisms include dysregulation of vascular endothelial growth factor and insulin-like growth factor 1. Furthermore, the gut microbiome may be impacted by other known risk factors for ROP, such as intermittent hypoxia and sepsis treated with antibiotics. This mini-review summarizes the literature supporting these proposed avenues, establishing a foundation to guide future studies.
Collapse
Affiliation(s)
- Jason Y Zhang
- Pritzker School of Medicine, University of Chicago, Chicago, Illinois; Department of Ophthalmology and Visual Science, University of Chicago, Chicago, Illinois
| | - Mark J Greenwald
- Department of Ophthalmology and Visual Science, University of Chicago, Chicago, Illinois
| | - Sarah H Rodriguez
- Department of Ophthalmology and Visual Science, University of Chicago, Chicago, Illinois.
| |
Collapse
|
14
|
Stanikova A, Jouza M, Bohosova J, Slaby O, Jabandziev P. Role of the microbiome in pathophysiology of necrotising enterocolitis in preterm neonates. BMJ Paediatr Open 2023; 7:e002172. [PMID: 37918941 PMCID: PMC10626796 DOI: 10.1136/bmjpo-2023-002172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 09/26/2023] [Indexed: 11/04/2023] Open
Abstract
Although necrotising enterocolitis (NEC) is a serious, life-threatening disease, improved neonatal care is increasing the number of survivors with NEC among extremely preterm neonates. Therapy is nevertheless mostly symptomatic and the mortality rate remains high, especially among neonates requiring surgery. Therefore, it is important to focus on preventing the disease and modifiable risk factors. NEC's pathophysiology is multifaceted, with key factors being immaturity of the immune and barrier protective mechanisms of the premature gut and exaggerated proinflammatory reaction to insults like gut hypoxia, enteral nutrition or microbial dysbiosis. The role of the intestinal microbiome in the pathophysiology of NEC has been a subject of research for many years, but to date no specific pathogen or type of dysbiosis has been connected with NEC development. This review assesses current knowledge as to the role of the intestinal microbiota in the pathophysiology of NEC and the possibilities for positively influencing it.
Collapse
Affiliation(s)
- Andrea Stanikova
- Department of Neonatology, University Hospital Brno, Brno, Czech Republic
- Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Martin Jouza
- Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Pediatrics, University Hospital Brno, Brno, Czech Republic
| | - Julia Bohosova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Ondrej Slaby
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Biology, University Hospital Brno, Brno, Czech Republic
| | - Petr Jabandziev
- Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Pediatrics, University Hospital Brno, Brno, Czech Republic
| |
Collapse
|
15
|
Chen X, Guo Q, Li YY, Song TY, Ge JQ. Metagenomic analysis fecal microbiota of dysentery-like diarrhoea in a pig farm using next-generation sequencing. Front Vet Sci 2023; 10:1257573. [PMID: 37915946 PMCID: PMC10616309 DOI: 10.3389/fvets.2023.1257573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 10/04/2023] [Indexed: 11/03/2023] Open
Abstract
Porcine enteric diseases including swine dysentery involves a wide range of possible aetiologies and seriously damages the intestine of pigs of all ages. Metagenomic next-generation sequencing is commonly used in research for detecting and analyzing pathogens. In this study, the feces of pigs from a commercial swine farm with dysentery-like diarrhea was collected and used for microbiota analysis by next-generation sequencing. While Brachyspira spp. was not detected in diarrheal pig fecal samples, indicating that the disease was not swine dysentery. The quantity of microbial population was extremely lowered, and the bacterial composition was altered with a reduction in the relative abundance of the probiotics organisms, Firmicutes and Bacteroidetes, with an increase in pathogens like Fusobacterium and Proteobacteria, in which the specific bacteria were identified at species-level. Viral pathogens, porcine circovirus type 2, porcine lymphotropic herpesviruses 1, and porcine mastadenovirus A were also detected at pretty low levels. Carbohydrate-active enzymes (CAZy) analysis indicated that the constitute of Firmicutes and Bacteroidete were also changed. Further, the Kyoto Encyclopedia of Genes and Genomes (KEGG) alignment analysis indicated that the microbiota of diarrheal pigs had a lower ability in utilizing energy sources but were enriched in multi-drug resistance pathways. Comprehensive Antibiotic Resistance Database (CARD) and Virulence Factors of Pathogenic Bacteria (VFDB) analysis indicated that genes for elfamycin and sulfonamide resistance and the iron uptake system were enriched in diarrheal pigs. This revealed potential bacterial infection and can guide antibiotic selection for treating dysentery. Overall, our data suggested that alterations in both the population and functional attributes of microbiota in diarrheal pigs with decreased probiotic and increased pathogenic microorganisms. These results will help elucidate the mechanism of dysentery-like diarrhea and the development of approaches to control the disease.
Collapse
Affiliation(s)
- Xi Chen
- Institute of Biotechnology, Fujian Academy of Agricultural Sciences, Fuzhou, Fujian, China
| | - Qing Guo
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, Fujian, China
| | - Ying-Ying Li
- Institute of Biotechnology, Fujian Academy of Agricultural Sciences, Fuzhou, Fujian, China
| | - Tie-Ying Song
- Institute of Biotechnology, Fujian Academy of Agricultural Sciences, Fuzhou, Fujian, China
| | - Jun-Qing Ge
- Institute of Biotechnology, Fujian Academy of Agricultural Sciences, Fuzhou, Fujian, China
| |
Collapse
|
16
|
Aires J, Ilhan ZE, Nicolas L, Ferraris L, Delannoy J, Bredel M, Chauvire-Drouard A, Barbut F, Rozé JC, Lepage P, Butel MJ. Occurrence of Neonatal Necrotizing Enterocolitis in Premature Neonates and Gut Microbiota: A Case-Control Prospective Multicenter Study. Microorganisms 2023; 11:2457. [PMID: 37894115 PMCID: PMC10609581 DOI: 10.3390/microorganisms11102457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 09/24/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is still one of the leading causes of neonatal death. The present study reports the data from a French case-control prospective multicenter study. METHODS A total of 146 preterm neonates (PNs) with or without NEC were included. Bacterial 16S rRNA gene sequencing was performed on stool samples (n = 103). Specific culture media were used to isolate Escherichia coli, Clostridium butyricum, and Clostridium neonatale, and strains were phenotypically characterized. RESULTS The gut microbiota of PNs was dominated by Firmicutes and Proteobacteria, and five enterotypes were identified. The microbiota composition was similar between NEC cases and PN controls. However, differences were observed in the relative abundance of Lactobacillus genus, which was significantly lower in the NEC group, whereas that of the Clostridium cluster III was significantly higher (p < 0.05). Within enterotypes, several phylotypes were significantly more abundant in NEC cases (p < 0.05). Regarding perinatal factors, a statistical association was found between the gut microbiota and cesarean delivery and antifungal therapy. In NEC cases and PN controls, the carriage rates and virulence genes of uropathogenic E. coli were equivalent based on culture. No correlation was found between E. coli, C. butyricum, and C. neonatale carriages, beta-lactam resistance, and antibiotic treatment. CONCLUSIONS At disease onset, our data support a microbiota dysbiosis between NEC and control infants at the genus level. In addition, it provides valuable information on bacterial antimicrobial susceptibility.
Collapse
Affiliation(s)
- Julio Aires
- Faculté de Pharmacie de Paris, Université Paris Cité, INSERM, UMR-S 1139 (3PHM), 75006 Paris, France (F.B.)
- FHU PREMA, Fighting Prematurity, 75014 Paris, France
| | - Zehra Esra Ilhan
- Micalis Institute, INRA, AgroParisTech, University Paris-Saclay, 91190 Paris, France; (Z.E.I.); (M.B.)
| | - Lancelot Nicolas
- Faculté de Pharmacie de Paris, Université Paris Cité, INSERM, UMR-S 1139 (3PHM), 75006 Paris, France (F.B.)
- FHU PREMA, Fighting Prematurity, 75014 Paris, France
| | - Laurent Ferraris
- Faculté de Pharmacie de Paris, Université Paris Cité, INSERM, UMR-S 1139 (3PHM), 75006 Paris, France (F.B.)
- FHU PREMA, Fighting Prematurity, 75014 Paris, France
| | - Johanne Delannoy
- Faculté de Pharmacie de Paris, Université Paris Cité, INSERM, UMR-S 1139 (3PHM), 75006 Paris, France (F.B.)
- FHU PREMA, Fighting Prematurity, 75014 Paris, France
| | - Maxime Bredel
- Micalis Institute, INRA, AgroParisTech, University Paris-Saclay, 91190 Paris, France; (Z.E.I.); (M.B.)
| | - Anne Chauvire-Drouard
- Centre d’Investigation Clinique CIC 1413, INSERM, CHU de Nantes, 44093 Nantes, France;
| | - Frédéric Barbut
- Faculté de Pharmacie de Paris, Université Paris Cité, INSERM, UMR-S 1139 (3PHM), 75006 Paris, France (F.B.)
- FHU PREMA, Fighting Prematurity, 75014 Paris, France
| | - Jean-Christophe Rozé
- Department of Neonatal Medicine, University Hospital of Nantes, 44093 Nantes, France;
| | - Patricia Lepage
- Micalis Institute, INRA, AgroParisTech, University Paris-Saclay, 91190 Paris, France; (Z.E.I.); (M.B.)
| | - Marie-José Butel
- Faculté de Pharmacie de Paris, Université Paris Cité, INSERM, UMR-S 1139 (3PHM), 75006 Paris, France (F.B.)
- FHU PREMA, Fighting Prematurity, 75014 Paris, France
| | | |
Collapse
|
17
|
Strobel KM, Juul SE, Hendrixson DT. Maternal Nutritional Status and the Microbiome across the Pregnancy and the Post-Partum Period. Microorganisms 2023; 11:1569. [PMID: 37375071 DOI: 10.3390/microorganisms11061569] [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: 05/15/2023] [Revised: 06/03/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Appropriate nutrition during pregnancy and the post-partum period is vital for both the mothers and their offspring. Both under- and over-nourished status may have important microbial implications on the maternal and infant gut microbiomes. Alterations in the microbiome can have implications for a person's risk of obesity and metabolic diseases. In this review, we examine alterations in the maternal gut, vaginal, placental, and milk microbiomes in the context of pre-pregnancy BMI, gestational weight gain, body composition, gestational diabetes, and maternal diet. We also investigate how the infant gut microbiome may be altered by these different parameters. Many of the microbial changes seen in under- and over-nourished states in birthing parents may result in long-term implications for the health of offspring. Differences in diet appear to be a major driver of the maternal and subsequently milk and offspring microbiomes. Further prospective longitudinal cohort studies are needed to examine nutrition and the microbiome to better understand its implications. Additionally, trials involving dietary interventions in child-bearing age adults should be explored to improve the mother and child's risks for metabolic diseases.
Collapse
Affiliation(s)
- Katie M Strobel
- Department of Pediatrics, University of Washington School of Medicine, 1959 NE Pacific St., Seattle, WA 98195, USA
| | - Sandra E Juul
- Department of Pediatrics, University of Washington School of Medicine, 1959 NE Pacific St., Seattle, WA 98195, USA
| | - David Taylor Hendrixson
- Department of Pediatrics, University of Washington School of Medicine, 1959 NE Pacific St., Seattle, WA 98195, USA
| |
Collapse
|
18
|
Rothers JL, Calton CM, Stepp JMB, Halpern MD. Enteral Feeding and Antibiotic Treatment Do Not Influence Increased Coefficient of Variation of Total Fecal Bile Acids in Necrotizing Enterocolitis. NEWBORN (CLARKSVILLE, MD.) 2023; 2:128-132. [PMID: 37559695 PMCID: PMC10411330 DOI: 10.5005/jp-journals-11002-0063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/11/2023]
Abstract
Introduction Necrotizing enterocolitis (NEC) is the most common gastrointestinal emergency in preterm infants. In animal models, the accumulation of ileal bile acids (BAs) is a crucial component of NEC pathophysiology. Recently, we showed that the coefficient of variation of total fecal BAs (CV-TBA) was elevated in infants who develop NEC compared to matched controls. However, neither the type of enteral nutrition nor antibiotic treatments-parameters that could potentially influence BA levels-were used to match pairs. Thus, we assessed the relationships between exposure to enteral feeding types and antibiotic treatments with NEC status and CV-TBA. Materials and methods Serial fecal samples were collected from 79 infants born with birth weight (BW) ≤1800 gm and estimated gestational age (EGA) ≤32 weeks; eighteen of these infants developed NEC. Total fecal BA levels (TBA) were determined using a commercially available enzyme cycling kit. Relationships between CV-TBA and dichotomous variables (NEC status, demographics, early exposure variables) were assessed by independent samples t-tests. Fisher's exact tests were used to assess relationships between NEC status and categorical variables. Results High values for CV-TBA levels perfectly predicted NEC status among infants in this study. However, feeding type and antibiotic usage did not drive this relationship. Conclusions As in previous studies, high values for the CV-TBA levels in the first weeks of life perfectly predicted NEC status among infants. Importantly, feeding type and antibiotic usage-previously identified risk factors for NEC-did not drive this relationship.
Collapse
Affiliation(s)
- Janet L Rothers
- BIO5 Institute Statistics Consulting Lab, University of Arizona, Tucson, Arizona, United States of America
| | - Christine M Calton
- Department of Pediatrics, University of Arizona College of Medicine, Tucson, Arizona, United States of America
| | - Jennifer MB Stepp
- Department of Family and Community Medicine, University of Arizona College of Medicine, Tucson, Arizona, United States of America
| | - Melissa D Halpern
- Department of Pediatrics, University of Arizona College of Medicine, Tucson, Arizona, United States of America
| |
Collapse
|
19
|
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: 4.0] [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
|
20
|
Xia J, Claud EC. Gut Microbiome-Brain Axis as an Explanation for the Risk of Poor Neurodevelopment Outcome in Preterm Infants with Necrotizing Enterocolitis. Microorganisms 2023; 11:1035. [PMID: 37110458 PMCID: PMC10142133 DOI: 10.3390/microorganisms11041035] [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: 03/13/2023] [Revised: 04/07/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Necrotizing Enterocolitis (NEC) is characterized by an inflammation of intestinal tissue that primarily affects premature infants. It is the most common and devastating gastrointestinal morbidity of prematurity, but beyond intestinal morbidity, this condition has also been associated with an increased risk of neurodevelopmental delays that persist beyond infancy. Prematurity, enteral feeding, bacterial colonization, and prolonged exposure to antibiotics are all risk factors that predispose preterm infants to NEC. Interestingly, these factors are all also associated with the gut microbiome. However, whether or not there is a connection between the microbiome and the risk of neurodevelopmental delays in infants after NEC is still an emerging area of research. Furthermore, how microbes in the gut could impact a distant organ such as the brain is also poorly understood. In this review, we discuss the current understanding of NEC and the role of the gut microbiome-brain axis in neurodevelopmental outcomes after NEC. Understanding the potential role of the microbiome in neurodevelopmental outcomes is important as the microbiome is modifiable and thus offers the hope of improved therapeutic options. We highlight the progress and limitations in this field. Insights into the gut microbiome-brain axis may offer potential therapeutic approaches to improve the long-term outcomes of premature infants.
Collapse
Affiliation(s)
- Jason Xia
- College of Liberal Arts and Sciences, University of Illinois Urbana-Champion, Champaign, IL 61801, USA
| | - Erika C. Claud
- Department of Pediatrics and Medicine, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
21
|
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: 7] [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
|
22
|
Duess JW, Sampah ME, Lopez CM, Tsuboi K, Scheese DJ, Sodhi CP, Hackam DJ. Necrotizing enterocolitis, gut microbes, and sepsis. Gut Microbes 2023; 15:2221470. [PMID: 37312412 PMCID: PMC10269420 DOI: 10.1080/19490976.2023.2221470] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 05/25/2023] [Indexed: 06/15/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a devastating disease in premature infants and the leading cause of death and disability from gastrointestinal disease in this vulnerable population. Although the pathophysiology of NEC remains incompletely understood, current thinking indicates that the disease develops in response to dietary and bacterial factors in the setting of a vulnerable host. As NEC progresses, intestinal perforation can result in serious infection with the development of overwhelming sepsis. In seeking to understand the mechanisms by which bacterial signaling on the intestinal epithelium can lead to NEC, we have shown that the gram-negative bacterial receptor toll-like receptor 4 is a critical regulator of NEC development, a finding that has been confirmed by many other groups. This review article provides recent findings on the interaction of microbial signaling, the immature immune system, intestinal ischemia, and systemic inflammation in the pathogenesis of NEC and the development of sepsis. We will also review promising therapeutic approaches that show efficacy in pre-clinical studies.
Collapse
Affiliation(s)
- Johannes W. Duess
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Maame E. Sampah
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Carla M. Lopez
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Koichi Tsuboi
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Daniel J. Scheese
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Chhinder P. Sodhi
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - David J. Hackam
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, USA
| |
Collapse
|
23
|
Kolba N, Cheng J, Jackson CD, Tako E. Intra-Amniotic Administration-An Emerging Method to Investigate Necrotizing Enterocolitis, In Vivo ( Gallus gallus). Nutrients 2022; 14:nu14224795. [PMID: 36432481 PMCID: PMC9696943 DOI: 10.3390/nu14224795] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/04/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a severe gastrointestinal disease in premature infants and a leading cause of death in neonates (1-7% in the US). NEC is caused by opportunistic bacteria, which cause gut dysbiosis and inflammation and ultimately result in intestinal necrosis. Previous studies have utilized the rodent and pig models to mimic NEC, whereas the current study uses the in vivo (Gallus gallus) intra-amniotic administration approach to investigate NEC. On incubation day 17, broiler chicken (Gallus gallus) viable embryos were injected intra-amniotically with 1 mL dextran sodium sulfate (DSS) in H2O. Four treatment groups (0.1%, 0.25%, 0.5%, and 0.75% DSS) and two controls (H2O/non-injected controls) were administered. We observed a significant increase in intestinal permeability and negative intestinal morphological changes, specifically, decreased villus surface area and goblet cell diameter in the 0.50% and 0.75% DSS groups. Furthermore, there was a significant increase in pathogenic bacterial (E. coli spp. and Klebsiella spp.) abundances in the 0.75% DSS group compared to the control groups, demonstrating cecal microbiota dysbiosis. These results demonstrate significant physiopathology of NEC and negative bacterial-host interactions within a premature gastrointestinal system. Our present study demonstrates a novel model of NEC through intra-amniotic administration to study the effects of NEC on intestinal functionality, morphology, and gut microbiota in vivo.
Collapse
Affiliation(s)
| | | | | | - Elad Tako
- Correspondence: ; Tel.: +1-607-255-0884
| |
Collapse
|
24
|
Cerdó T, García-Santos JA, Rodríguez-Pöhnlein A, García-Ricobaraza M, Nieto-Ruíz A, G. Bermúdez M, Campoy C. Impact of Total Parenteral Nutrition on Gut Microbiota in Pediatric Population Suffering Intestinal Disorders. Nutrients 2022; 14:4691. [PMID: 36364953 PMCID: PMC9658482 DOI: 10.3390/nu14214691] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 08/10/2023] Open
Abstract
Parenteral nutrition (PN) is a life-saving therapy providing nutritional support in patients with digestive tract complications, particularly in preterm neonates due to their gut immaturity during the first postnatal weeks. Despite this, PN can also result in several gastrointestinal complications that are the cause or consequence of gut mucosal atrophy and gut microbiota dysbiosis, which may further aggravate gastrointestinal disorders. Consequently, the use of PN presents many unique challenges, notably in terms of the potential role of the gut microbiota on the functional and clinical outcomes associated with the long-term use of PN. In this review, we synthesize the current evidence on the effects of PN on gut microbiome in infants and children suffering from diverse gastrointestinal diseases, including necrotizing enterocolitis (NEC), short bowel syndrome (SBS) and subsequent intestinal failure, liver disease and inflammatory bowel disease (IBD). Moreover, we discuss the potential use of pre-, pro- and/or synbiotics as promising therapeutic strategies to reduce the risk of severe gastrointestinal disorders and mortality. The findings discussed here highlight the need for more well-designed studies, and harmonize the methods and its interpretation, which are critical to better understand the role of the gut microbiota in PN-related diseases and the development of efficient and personalized approaches based on pro- and/or prebiotics.
Collapse
Affiliation(s)
- Tomás Cerdó
- Maimonides Institute for Research in Biomedicine of Córdoba (IMIBIC), Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain
| | - José Antonio García-Santos
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria Ibs-GRANADA, Health Sciences Technological Park, 18012 Granada, Spain
| | - Anna Rodríguez-Pöhnlein
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria Ibs-GRANADA, Health Sciences Technological Park, 18012 Granada, Spain
| | - María García-Ricobaraza
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria Ibs-GRANADA, Health Sciences Technological Park, 18012 Granada, Spain
| | - Ana Nieto-Ruíz
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria Ibs-GRANADA, Health Sciences Technological Park, 18012 Granada, Spain
| | - Mercedes G. Bermúdez
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria Ibs-GRANADA, Health Sciences Technological Park, 18012 Granada, Spain
| | - Cristina Campoy
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria Ibs-GRANADA, Health Sciences Technological Park, 18012 Granada, Spain
- Spanish Network of Biomedical Research in Epidemiology and Public Health (CIBERESP), Granada’s Node, Carlos III Health Institute, Avda. Monforte de Lemos 5, 28028 Madrid, Spain
| |
Collapse
|
25
|
Moschino L, Verlato G, Duci M, Cavicchiolo ME, Guiducci S, Stocchero M, Giordano G, Fascetti Leon F, Baraldi E. The Metabolome and the Gut Microbiota for the Prediction of Necrotizing Enterocolitis and Spontaneous Intestinal Perforation: A Systematic Review. Nutrients 2022; 14:nu14183859. [PMID: 36145235 PMCID: PMC9506026 DOI: 10.3390/nu14183859] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/13/2022] [Accepted: 09/13/2022] [Indexed: 11/26/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is the most devastating gastrointestinal emergency in preterm neonates. Research on early predictive biomarkers is fundamental. This is a systematic review of studies applying untargeted metabolomics and gut microbiota analysis to evaluate the differences between neonates affected by NEC (Bell’s stage II or III), and/or by spontaneous intestinal perforation (SIP) versus healthy controls. Five studies applying metabolomics (43 cases, 95 preterm controls) and 20 applying gut microbiota analysis (254 cases, 651 preterm controls, 22 term controls) were selected. Metabolomic studies utilized NMR spectroscopy or mass spectrometry. An early urinary alanine/histidine ratio >4 showed good sensitivity and predictive value for NEC in one study. Samples collected in proximity to NEC diagnosis demonstrated variable pathways potentially related to NEC. In studies applying untargeted gut microbiota analysis, the sequencing of the V3−V4 or V3 to V5 regions of the 16S rRNA was the most used technique. At phylum level, NEC specimens were characterized by increased relative abundance of Proteobacteria compared to controls. At genus level, pre-NEC samples were characterized by a lack or decreased abundance of Bifidobacterium. Finally, at the species level Bacteroides dorei, Clostridium perfringens and perfringens-like strains dominated early NEC specimens, whereas Clostridium butyricum, neonatale and Propionibacterium acnei those at disease diagnosis. Six studies found a lower Shannon diversity index in cases than controls. A clear separation of cases from controls emerged based on UniFrac metrics in five out of seven studies. Importantly, no studies compared NEC versus SIP. Untargeted metabolomics and gut microbiota analysis are interrelated strategies to investigate NEC pathophysiology and identify potential biomarkers. Expression of quantitative measurements, data sharing via biorepositories and validation studies are fundamental to guarantee consistent comparison of results.
Collapse
Affiliation(s)
- Laura Moschino
- Neonatal Intensive Care Unit, Department of Women’s and Children’s Health, Padova University Hospital, 35128 Padova, Italy
- Institute of Paediatric Research, Città della Speranza, Laboratory of Mass Spectrometry and Metabolomics, 35127 Padova, Italy
- Correspondence: ; Tel.: +39-049-821-3548
| | - Giovanna Verlato
- Neonatal Intensive Care Unit, Department of Women’s and Children’s Health, Padova University Hospital, 35128 Padova, Italy
| | - Miriam Duci
- Paediatric Surgery, Department of Women’s and Children’s Health, Padova University Hospital, 35128 Padova, Italy
| | - Maria Elena Cavicchiolo
- Neonatal Intensive Care Unit, Department of Women’s and Children’s Health, Padova University Hospital, 35128 Padova, Italy
| | - Silvia Guiducci
- Neonatal Intensive Care Unit, Department of Women’s and Children’s Health, Padova University Hospital, 35128 Padova, Italy
| | - Matteo Stocchero
- Institute of Paediatric Research, Città della Speranza, Laboratory of Mass Spectrometry and Metabolomics, 35127 Padova, Italy
- Laboratory of Mass Spectrometry and Metabolomics, Department of Women’s and Children’s Health, Padova University Hospital, 35128 Padova, Italy
| | - Giuseppe Giordano
- Institute of Paediatric Research, Città della Speranza, Laboratory of Mass Spectrometry and Metabolomics, 35127 Padova, Italy
- Laboratory of Mass Spectrometry and Metabolomics, Department of Women’s and Children’s Health, Padova University Hospital, 35128 Padova, Italy
| | - Francesco Fascetti Leon
- Paediatric Surgery, Department of Women’s and Children’s Health, Padova University Hospital, 35128 Padova, Italy
| | - Eugenio Baraldi
- Neonatal Intensive Care Unit, Department of Women’s and Children’s Health, Padova University Hospital, 35128 Padova, Italy
- Institute of Paediatric Research, Città della Speranza, Laboratory of Mass Spectrometry and Metabolomics, 35127 Padova, Italy
| |
Collapse
|
26
|
Dai D, Dai F, Chen J, Jin M, Li M, Hu D, Liu Z, Zhang Z, Xu F, Chen WH. Integrated multi-omics reveal important roles of gut contents in intestinal ischemia–reperfusion induced injuries in rats. Commun Biol 2022; 5:938. [PMID: 36085351 PMCID: PMC9463172 DOI: 10.1038/s42003-022-03887-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 08/08/2022] [Indexed: 12/13/2022] Open
Abstract
Intestinal ischemia–reperfusion (IIR) is a life-threatening clinical event with damaging signals whose origin and contents are unclear. Here we observe that IIR significantly affect the metabolic profiles of most organs by unbiased organ-wide metabolic analysis of gut contents, blood, and fifteen organs in rats (n = 29). Remarkably, correlations between gut content metabolic profiles and those of other organs are the most significant. Gut contents are also the only ones to show dynamic correlations during IIR. Additionally, according to targeted metabolomics analysis, several neurotransmitters are considerably altered in the gut during IIR, and displayed noteworthy correlations with remote organs. Likewise, metagenomics analysis (n = 35) confirm the effects of IIR on gut microbiota, and identify key species fundamental to the changes in gut metabolites, particularly neurotransmitters. Our multi-omics results establish key roles of gut contents in IIR induced remote injury and provide clues for future exploration. Die Dai et al. evaluate changes in the metabolomic and gut microbiome in response to experimental intestinal ischemia reperfusion (IIR) injury in rats. Their results provide further insight into how gut contents contribute to widespread injury in IIR patients.
Collapse
|
27
|
Subramanian S, Geng H, Du C, Chou PM, Bu HF, Wang X, Swaminathan S, Tan SC, Ridlon JM, De Plaen IG, Tan XD. Feeding mode influences dynamic gut microbiota signatures and affects susceptibility to anti-CD3 mAb-induced intestinal injury in neonatal mice. Am J Physiol Gastrointest Liver Physiol 2022; 323:G205-G218. [PMID: 35819158 PMCID: PMC9394775 DOI: 10.1152/ajpgi.00337.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 07/01/2022] [Accepted: 07/02/2022] [Indexed: 01/31/2023]
Abstract
Feeding modes influence the gut microbiome, immune system, and intestinal barrier homeostasis in neonates; how feeding modes impact susceptibility to neonatal gastrointestinal (GI) diseases is still uncertain. Here, we investigated the impact of dam feeding (DF) and formula feeding (FF) on features of the gut microbiome and physiological inflammation during the first 2 days of postnatal development and on the susceptibility to intestinal injury related to the inflammatory state in neonatal mouse pups. 16S rRNA sequencing data revealed microbiome changes, lower α-diversity, and a distinct pattern of β-diversity including expansion of f_Enterobacteriaceae and f_Enterococcaceae in the ileum of FF pups compared with DF pups by postnatal day (P)2. Together with gut dysbiosis, the FF cohort also had greater ileal mucosa physiological inflammatory activity compared with DF pups by P2 but maintained normal histological features. Interestingly, FF but not DF mouse pups developed necrotizing enterocolitis (NEC)-like intestinal injury within 24 h after anti-CD3 mAb treatment, suggesting that FF influences the susceptibility to intestinal injury in neonates. We further found that NEC-like incidence in anti-CD3 mAb-treated FF neonatal pups was attenuated by antibiotic treatment. Collectively, our data suggest that FF predisposes mouse pups to anti-CD3 mAb-induced intestinal injury due to abnormal f_Enterobacteriaceae and f_Enterococcaceae colonization. These findings advance our understanding of FF-associated microbial colonization and intestinal inflammation, which may help inform the development of new therapeutic approaches to GI diseases like NEC in infants.NEW & NOTEWORTHY This report shows that a feeding mode profoundly affects gut colonization in neonatal mice. Furthermore, our results demonstrate that formula feeding predisposes mouse pups to anti-CD3 mAb-induced necrotizing enterocolitis (NEC)-like intestinal injury upon inadequate microbial colonization. The study suggests the role of the combined presence of formula feeding-associated dysbiosis and mucosal inflammation in the pathogenesis of NEC and provides a new mouse model to study this disease.
Collapse
Affiliation(s)
- Saravanan Subramanian
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Center for Intestinal and Liver Inflammation Research, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Hua Geng
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Center for Intestinal and Liver Inflammation Research, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Chao Du
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Center for Intestinal and Liver Inflammation Research, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Pauline M Chou
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Heng-Fu Bu
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Center for Intestinal and Liver Inflammation Research, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Xiao Wang
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Center for Intestinal and Liver Inflammation Research, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Suchitra Swaminathan
- Division of Rheumatology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Stephanie C Tan
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Center for Intestinal and Liver Inflammation Research, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Jason M Ridlon
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, Illinois
| | - Isabelle G De Plaen
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Division of Neonatology, Department of Pediatrics, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Xiao-Di Tan
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Center for Intestinal and Liver Inflammation Research, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Department of Research and Development, Jesse Brown Department of Veterans Affairs Medical Center, Chicago, Illinois
| |
Collapse
|
28
|
Exploring the Potential of Human Milk and Formula Milk on Infants’ Gut and Health. Nutrients 2022; 14:nu14173554. [PMID: 36079814 PMCID: PMC9460722 DOI: 10.3390/nu14173554] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 11/21/2022] Open
Abstract
Early-life gut microbiota plays a role in determining the health and risk of developing diseases in later life. Various perinatal factors have been shown to contribute to the development and establishment of infant gut microbiota. One of the important factors influencing the infant gut microbial colonization and composition is the mode of infant feeding. While infant formula milk has been designed to resemble human milk as much as possible, the gut microbiome of infants who receive formula milk differs from that of infants who are fed human milk. A diverse microbial population in human milk and the microbes seed the infant gut microbiome. Human milk contains nutritional components that promote infant growth and bioactive components, such as human milk oligosaccharides, lactoferrin, and immunoglobulins, which contribute to immunological development. In an attempt to encourage the formation of a healthy gut microbiome comparable to that of a breastfed infant, manufacturers often supplement infant formula with prebiotics or probiotics, which are known to have a bifidogenic effect and can modulate the immune system. This review aims to elucidate the roles of human milk and formula milk on infants’ gut and health.
Collapse
|
29
|
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:antibiotics11020237. [PMID: 35203839 PMCID: PMC8868158 DOI: 10.3390/antibiotics11020237] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [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
- Correspondence:
| | - 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
|
30
|
The Microbiota-Gut Axis in Premature Infants: Physio-Pathological Implications. Cells 2022; 11:cells11030379. [PMID: 35159189 PMCID: PMC8834399 DOI: 10.3390/cells11030379] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/22/2021] [Accepted: 01/17/2022] [Indexed: 11/16/2022] Open
Abstract
Intriguing evidence is emerging in regard to the influence of gut microbiota composition and function on host health from the very early stages of life. The development of the saprophytic microflora is conditioned by several factors in infants, and peculiarities have been found for babies born prematurely. This population is particularly exposed to a high risk of infection, postnatal antibiotic treatment, feeding difficulties and neurodevelopmental disabilities. To date, there is still a wide gap in understanding all the determinants and the mechanism behind microbiota disruption and its influence in the development of the most common complications of premature infants. A large body of evidence has emerged during the last decades showing the existence of a bidirectional communication axis involving the gut microbiota, the gut and the brain, defined as the microbiota–gut–brain axis. In this context, given that very few data are available to demonstrate the correlation between microbiota dysbiosis and neurodevelopmental disorders in preterm infants, increasing interest has arisen to better understand the impact of the microbiota–gut–brain axis on the clinical outcomes of premature infants and to clarify how this may lead to alternative preventive, diagnostic and therapeutic strategies. In this review, we explored the current evidence regarding microbiota development in premature infants, focusing on the effects of delivery mode, type of feeding, environmental factors and possible influence of the microbiota–gut–brain axis on preterm clinical outcomes during their hospital stay and on their health status later in life.
Collapse
|
31
|
Ahmed S, Travis SD, Díaz-Bahamonde FV, Porter DDL, Henry SN, Mykins J, Ravipati A, Booker A, Ju J, Ding H, Ramesh AK, Pickrell AM, Wang M, LaConte S, Howell BR, Yuan L, Morton PD. Early Influences of Microbiota on White Matter Development in Germ-Free Piglets. Front Cell Neurosci 2022; 15:807170. [PMID: 35027884 PMCID: PMC8751630 DOI: 10.3389/fncel.2021.807170] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 11/29/2021] [Indexed: 11/13/2022] Open
Abstract
Abnormalities in the prefrontal cortex (PFC), as well as the underlying white matter (WM) tracts, lie at the intersection of many neurodevelopmental disorders. The influence of microorganisms on brain development has recently been brought into the clinical and research spotlight as alterations in commensal microbiota are implicated in such disorders, including autism spectrum disorders, schizophrenia, depression, and anxiety via the gut-brain axis. In addition, gut dysbiosis is common in preterm birth patients who often display diffuse WM injury and delayed WM maturation in critical tracts including those within the PFC and corpus callosum. Microbial colonization of the gut aligns with ongoing postnatal processes of oligodendrogenesis and the peak of brain myelination in humans; however, the influence of microbiota on gyral WM development remains elusive. Here, we develop and validate a neonatal germ-free swine model to address these issues, as piglets share key similarities in WM volume, developmental trajectories, and distribution to humans. We find significant region-specific reductions, and sexually dimorphic trends, in WM volume, oligodendrogenesis, and mature oligodendrocyte numbers in germ-free piglets during a key postnatal epoch of myelination. Our findings indicate that microbiota plays a critical role in promoting WM development during early life when the brain is vulnerable to environmental insults that can result in an array of disabilities manifesting later in life.
Collapse
Affiliation(s)
- Sadia Ahmed
- Graduate Studies in Biomedical and Veterinary Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Sierrah D Travis
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Francisca V Díaz-Bahamonde
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Demisha D L Porter
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States.,Virginia Tech Graduate Program in Translational Biology, Medicine and Health, Virginia Tech, Roanoke, VA, United States
| | - Sara N Henry
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Julia Mykins
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Aditya Ravipati
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Aryn Booker
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Jing Ju
- Graduate Studies in Biomedical and Veterinary Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Hanzhang Ding
- Virginia Tech Graduate Program in Translational Biology, Medicine and Health, Virginia Tech, Roanoke, VA, United States
| | - Ashwin K Ramesh
- Graduate Studies in Biomedical and Veterinary Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States.,Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States.,Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, United States
| | - Alicia M Pickrell
- School of Neuroscience, Virginia Tech, Blacksburg, VA, United States
| | - Maosen Wang
- Fralin Biomedical Research Institute at Virginia Tech Carilion (VTC), Virginia Tech, Roanoke, VA, United States
| | - Stephen LaConte
- Fralin Biomedical Research Institute at Virginia Tech Carilion (VTC), Virginia Tech, Roanoke, VA, United States.,Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, United States
| | - Brittany R Howell
- Fralin Biomedical Research Institute at Virginia Tech Carilion (VTC), Virginia Tech, Roanoke, VA, United States.,Department of Human Development and Family Science, Virginia Tech, Roanoke, VA, United States
| | - Lijuan Yuan
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Paul D Morton
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
32
|
Verma J, Sankar MJ, Atmakuri K, Agarwal R, Das B. Gut microbiome dysbiosis in neonatal sepsis. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 192:125-147. [DOI: 10.1016/bs.pmbts.2022.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
33
|
Casaburi G, Wei J, Kazi S, Liu J, Wang K, Tao GZ, Lin PY, Dunn JCY, Henrick BM, Frese SA, Sylvester KG. Metabolic model of necrotizing enterocolitis in the premature newborn gut resulting from enteric dysbiosis. Front Pediatr 2022; 10:893059. [PMID: 36081629 PMCID: PMC9445129 DOI: 10.3389/fped.2022.893059] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/24/2022] [Indexed: 11/29/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a leading cause of premature newborn morbidity and mortality. The clinical features of NEC consistently include prematurity, gut dysbiosis and enteral inflammation, yet the pathogenesis remains obscure. Herein we combine metagenomics and targeted metabolomics, with functional in vivo and in vitro assessment, to define a novel molecular mechanism of NEC. One thousand six hundred and forty seven publicly available metagenomics datasets were analyzed (NEC = 245; healthy = 1,402) using artificial intelligence methodologies. Targeted metabolomic profiling was used to quantify the concentration of specified fecal metabolites at NEC onset (n = 8), during recovery (n = 6), and in age matched controls (n = 10). Toxicity assays of discovered metabolites were performed in vivo in mice and in vitro using human intestinal epithelial cells. Metagenomic and targeted metabolomic analyses revealed significant differences in pyruvate fermentation pathways and associated intermediates. Notably, the short chain fatty acid formate was elevated in the stool of NEC patients at disease onset (P = 0.005) dissipated during recovery (P = 0.02) and positively correlated with degree of intestinal injury (r 2 = 0.86). In vitro, formate caused enterocyte cytotoxicity in human cells through necroptosis (P < 0.01). In vivo, luminal formate caused significant dose and development dependent NEC-like injury in newborn mice. Enterobacter cloacae and Klebsiella pneumoniae were the most discriminatory taxa related to NEC dysbiosis and increased formate production. Together, these data suggest a novel biochemical mechanism of NEC through the microbial production of formate. Clinical efforts to prevent NEC should focus on reducing the functional consequences of newborn gut dysbiosis associated metabolic pathways.
Collapse
Affiliation(s)
| | - Jingjing Wei
- Department of Surgery, Stanford University, Stanford, CA, United States.,Department of Pediatrics, Shanxi Medical University, Taiyuan, China
| | - Sufyan Kazi
- Evolve Biosystems, Inc., Davis, CA, United States
| | - Junlin Liu
- Department of Surgery, Stanford University, Stanford, CA, United States.,Department of General Surgery, The People's Hospital of Liuyang City, Liuyang, China
| | - Kewei Wang
- Department of Surgery, Stanford University, Stanford, CA, United States.,Department of Gastrointestinal Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Guo-Zhong Tao
- Department of Surgery, Stanford University, Stanford, CA, United States
| | - Po-Yu Lin
- Department of Surgery, Stanford University, Stanford, CA, United States
| | - James C Y Dunn
- Department of Surgery, Stanford University, Stanford, CA, United States
| | - Bethany M Henrick
- Evolve Biosystems, Inc., Davis, CA, United States.,Department of Food Science and Technology, University of Nebraska, Lincoln, NE, United States
| | - Steven A Frese
- Evolve Biosystems, Inc., Davis, CA, United States.,Department of Food Science and Technology, University of Nebraska, Lincoln, NE, United States.,Department of Nutrition, University of Nevada Reno, Reno, NV, United States
| | - Karl G Sylvester
- Department of Surgery, Stanford University, Stanford, CA, United States
| |
Collapse
|
34
|
The microbiome, guard or threat to infant health. Trends Mol Med 2021; 27:1175-1186. [PMID: 34518093 DOI: 10.1016/j.molmed.2021.08.002] [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: 06/15/2021] [Revised: 07/29/2021] [Accepted: 08/11/2021] [Indexed: 11/23/2022]
Abstract
Despite improvements in survival for very low birthweight (VLBW) premature infants, there continues to be significant morbidity for these infants at remarkable cost to the healthcare system. Concurrent development of the preterm infant intestine alongside the gut microbiome in the clinical setting rather than in the protected in utero environment where it would usually occur creates significant vulnerabilities for the infant's immature intestine and immune system, resulting in devastating illness and neurological injury. However, the microbiome also has the capacity to promote healthy development. Studies of parallel gut microbiome and preterm infant development have given key insight into the impact of the microbiome on intestinal as well as neural development and may provide potential therapeutic targets to prevent preterm infant morbidities.
Collapse
|
35
|
Heida FH, Kooi EMW, Wagner J, Nguyen TY, Hulscher JBF, van Zoonen AGJF, Bos AF, Harmsen HJM, de Goffau MC. Weight shapes the intestinal microbiome in preterm infants: results of a prospective observational study. BMC Microbiol 2021; 21:219. [PMID: 34289818 PMCID: PMC8293572 DOI: 10.1186/s12866-021-02279-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 06/28/2021] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND The intestinal microbiome in preterm infants differs markedly from term infants. It is unclear whether the microbiome develops over time according to infant specific factors. METHODS We analysed (clinical) metadata - to identify the main factors influencing the microbiome composition development - and the first meconium and faecal samples til the 4th week via 16 S rRNA amplican sequencing. RESULTS We included 41 infants (gestational age 25-30 weeks; birth weight 430-990 g. Birth via Caesarean section (CS) was associated with placental insufficiency during pregnancy and lower BW. In meconium samples and in samples from weeks 2 and 3 the abundance of Escherichia and Bacteroides (maternal faecal representatives) were associated with vaginal delivery while Staphylococcus (skin microbiome representative) was associated with CS. Secondly, irrespective of the week of sampling or the mode of birth, a transition was observed as children children gradually increased in weight from a microbiome dominated by Staphylococcus (Bacilli) towards a microbiome dominated by Enterobacteriaceae (Gammaproteobacteria). CONCLUSIONS Our data show that the mode of delivery affects the meconium microbiome composition. They also suggest that the weight of the infant at the time of sampling is a better predictor for the stage of progression of the intestinal microbiome development/maturation than postconceptional age as it less confounded by various infant-specific factors.
Collapse
Affiliation(s)
- Fardou H Heida
- Division of Obstetrics & Gynecology, Isala Klinieken, University of Groningen, Zwolle, the Netherlands. .,Division of Pediatric Surgery Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| | - Elisabeth M W Kooi
- Division of Neonatology Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Josef Wagner
- Victorian Infectious Diseases Reference Laboratory, Peter Doherty Institute for Infection and Immunity, Royal Melbourne Hospital, Melbourne, Australia
| | - Thi-Yen Nguyen
- Division of Pediatric Surgery Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jan B F Hulscher
- Division of Pediatric Surgery Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Anne G J F van Zoonen
- Division of Pediatric Surgery Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Arend F Bos
- Division of Neonatology Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Hermie J M Harmsen
- Division of Microbiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Marcus C de Goffau
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands.,Parasites and Microboes, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| |
Collapse
|
36
|
Buffet-Bataillon S, Bellanger A, Boudry G, Gangneux JP, Yverneau M, Beuchée A, Blat S, Le Huërou-Luron I. New Insights Into Microbiota Modulation-Based Nutritional Interventions for Neurodevelopmental Outcomes in Preterm Infants. Front Microbiol 2021; 12:676622. [PMID: 34177860 PMCID: PMC8232935 DOI: 10.3389/fmicb.2021.676622] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/12/2021] [Indexed: 12/19/2022] Open
Abstract
Gut microbiota and the central nervous system have parallel developmental windows during pre and post-natal life. Increasing evidences suggest that intestinal dysbiosis in preterm infants predisposes the neonate to adverse neurological outcomes later in life. Understanding the link between gut microbiota colonization and brain development to tailor therapies aimed at optimizing initial colonization and microbiota development are promising strategies to warrant adequate brain development and enhance neurological outcomes in preterm infants. Breast-feeding has been associated with both adequate cognitive development and healthy microbiota in preterms. Infant formula are industrially produced substitutes for infant nutrition that do not completely recapitulate breast-feeding benefices and could be largely improved by the understanding of the role of breast milk components upon gut microbiota. In this review, we will first discuss the nutritional and bioactive component information on breast milk composition and its contribution to the assembly of the neonatal gut microbiota in preterms. We will then discuss the emerging pathways connecting the gut microbiota and brain development. Finally, we will discuss the promising microbiota modulation-based nutritional interventions (including probiotic and prebiotic supplementation of infant formula and maternal nutrition) for improving neurodevelopmental outcomes.
Collapse
Affiliation(s)
- Sylvie Buffet-Bataillon
- Institut NuMeCan, INRAE, INSERM, Univ Rennes, Saint-Gilles, France
- Department of Clinical Microbiology, CHU Rennes, Rennes, France
| | - Amandine Bellanger
- Institut NuMeCan, INRAE, INSERM, Univ Rennes, Saint-Gilles, France
- Department of Pediatrics-Neonatology, CHU Rennes, Rennes, France
| | - Gaelle Boudry
- Institut NuMeCan, INRAE, INSERM, Univ Rennes, Saint-Gilles, France
| | | | | | - Alain Beuchée
- Department of Pediatrics-Neonatology, Univ Rennes, CHU Rennes, LTSI-UMR 1099, Rennes, France
| | - Sophie Blat
- Institut NuMeCan, INRAE, INSERM, Univ Rennes, Saint-Gilles, France
| | | |
Collapse
|
37
|
Huang Q, Lu S, Zhu Y, Wei B, Chen Y, Bai F. Bacterial endotoxin-induced maternal inflammation leads to fetal intestinal injury and affects microbial colonization in the neonatal period. J Matern Fetal Neonatal Med 2021; 35:6917-6927. [PMID: 34058958 DOI: 10.1080/14767058.2021.1931101] [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: 10/21/2022]
Abstract
BACKGROUND Intraamniotic infection is associated with an increased risk of multiple adverse outcomes in offspring, especially neonatal necrotizing enterocolitis (NEC), which is one of the serious gastrointestinal diseases in neonates. However, the underlying mechanism remains undefined. We hypothesize that bacterial endotoxin-induced maternal inflammation causes intestinal injury in offspring, thereby affecting the composition of the intestinal microbiome. METHODS Pregnant Sprague Dawley rats were received intraperitoneal injections with 700 μg/kg lipopolysaccharide (LPS, which was the same as bacterial endotoxin) or saline at 15 days of gestation. Pups were allowed to deliver naturally and euthanized at days 0, 3 and 7 after birth. Intestinal tissue and feces samples from offspring were collected to evaluate the effects of maternal inflammation on intestinal flora colonization and intestinal mucosal development. RESULTS Significant intestinal injury of the offspring induced by prenatal LPS exposure was observed on day 0 and 3 after birth. In addition, prenatal LPS exposure also induced significant changes in the intestinal microbiome of the offspring with a significant increase in Proteobacteria (Escherichia-Shigella) and a decrease in Firmicutes at 7 days after birth. CONCLUSIONS Thus, our findings suggest that LPS-induced maternal inflammation induces intestinal injury in offspring and subsequently leads to NEC-like changes in the composition of the intestinal microbiome.
Collapse
Affiliation(s)
- Qingmei Huang
- Department of Pediatrics, The Second Affiliated Hospital of Guangxi Medical University, Guangxi, China
| | - Siliang Lu
- Department of Pediatrics, The Second Affiliated Hospital of Guangxi Medical University, Guangxi, China
| | - Yunlei Zhu
- Department of Pediatrics, The Second Affiliated Hospital of Guangxi Medical University, Guangxi, China
| | - Bingmei Wei
- Department of Pediatrics, The Second Affiliated Hospital of Guangxi Medical University, Guangxi, China
| | - Yujun Chen
- Department of Pediatrics, The Second Affiliated Hospital of Guangxi Medical University, Guangxi, China
| | - Faming Bai
- Department of Pediatrics, The Second Affiliated Hospital of Guangxi Medical University, Guangxi, China
| |
Collapse
|
38
|
Sampah MES, Hackam DJ. Prenatal Immunity and Influences on Necrotizing Enterocolitis and Associated Neonatal Disorders. Front Immunol 2021; 12:650709. [PMID: 33968047 PMCID: PMC8097145 DOI: 10.3389/fimmu.2021.650709] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 04/06/2021] [Indexed: 12/14/2022] Open
Abstract
Prior to birth, the neonate has limited exposure to pathogens. The transition from the intra-uterine to the postnatal environment initiates a series of complex interactions between the newborn host and a variety of potential pathogens that persist over the first few weeks of life. This transition is particularly complex in the case of the premature and very low birth weight infant, who may be susceptible to many disorders as a result of an immature and underdeveloped immune system. Chief amongst these disorders is necrotizing enterocolitis (NEC), an acute inflammatory disorder that leads to necrosis of the intestine, and which can affect multiple systems and have the potential to result in long term effects if the infant is to survive. Here, we examine what is known about the interplay of the immune system with the maternal uterine environment, microbes, nutritional and other factors in the pathogenesis of neonatal pathologies such as NEC, while also taking into consideration the effects on the long-term health of affected children.
Collapse
Affiliation(s)
| | - David J. Hackam
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine , Baltimore, MD, United States
| |
Collapse
|
39
|
Moore RE, Xu LL, Townsend SD. Prospecting Human Milk Oligosaccharides as a Defense Against Viral Infections. ACS Infect Dis 2021; 7:254-263. [PMID: 33470804 DOI: 10.1021/acsinfecdis.0c00807] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In addition to providing maximal nutritional value for neonatal growth and development, human milk functions as an early defense mechanism against invading pathogens. Human milk oligosaccharides (HMOs), which are abundant in human milk, are a diverse group of heterogeneous carbohydrates with wide ranging protective effects. In addition to promoting the colonization of beneficial intestinal flora, HMOs serve as decoy receptors, effectively blocking the attachment of pathogenic bacteria. HMOs also function as bacteriostatic agents, inhibiting the growth of gram-positive bacteria. Based on this precedence, an emerging area in the field has focused on characterizing the antiviral properties of HMOs. Indeed, HMOs have been evaluated as antiviral agents, with many possessing activity against life-threatening infections. This targeted review provides insight into the known glycan-binding interactions between select HMOs and influenza, rotavirus, respiratory syncytial virus, human immunodeficiency virus, and norovirus. Additionally, we review the role of HMOs in preventing necrotizing enterocolitis, an intestinal disease linked to viral infections. We close with a discussion of what is known broadly regarding human milk oligosaccharides and their interactions with coronaviruses.
Collapse
Affiliation(s)
- Rebecca E. Moore
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Lianyan L. Xu
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Steven D. Townsend
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37212, United States
- Vanderbilt Microbiome Initiative, Vanderbilt University, Nashville, Tennessee 37212, United States
| |
Collapse
|
40
|
Venkatraman A, Yu W, Nitkin C, Sampath V. Intestinal Stem Cell Development in the Neonatal Gut: Pathways Regulating Development and Relevance to Necrotizing Enterocolitis. Cells 2021; 10:cells10020312. [PMID: 33546361 PMCID: PMC7913590 DOI: 10.3390/cells10020312] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/21/2021] [Accepted: 01/29/2021] [Indexed: 12/15/2022] Open
Abstract
The intestine is extremely dynamic and the epithelial cells that line the intestine get replaced every 3–5 days by highly proliferative intestinal stem cells (ISCs). The instructions for ISCs to self-renew or to differentiate come as cues from their surrounding microenvironment or their niche. A small number of evolutionarily conserved signaling pathways act as a critical regulator of the stem cells in the adult intestine, and these pathways are well characterized. However, the mechanisms, nutritional, and environmental signals that help establish the stem cell niche in the neonatal intestine are less studied. Deciphering the key signaling pathways that regulate the development and maintenance of the stem cells is particularly important to understanding how the intestine regenerates from necrotizing enterocolitis, a devastating disease in newborn infants characterized by inflammation, tissues necrosis, and stem cell injury. In this review, we piece together current knowledge on morphogenetic and immune pathways that regulate intestinal stem cell in neonates and highlight how the cross talk among these pathways affect tissue regeneration. We further discuss how these key pathways are perturbed in NEC and review the scientific knowledge relating to options for stem cell therapy in NEC gleaned from pre-clinical experimental models of NEC.
Collapse
|
41
|
van Best N, Trepels-Kottek S, Savelkoul P, Orlikowsky T, Hornef MW, Penders J. Influence of probiotic supplementation on the developing microbiota in human preterm neonates. Gut Microbes 2020; 12:1-16. [PMID: 33095113 PMCID: PMC7588225 DOI: 10.1080/19490976.2020.1826747] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Oral administration of probiotic bacteria to preterm neonates has been recommended to prevent the development of necrotizing enterocolitis (NEC). The influence of probiotics on the endogenous microbiome, however, has remained incompletely understood. STUDY DESIGN & METHODS Here, we performed an observational study including 80 preterm neonates born at a gestational age <32-weeks to characterize the persistence of probiotic bacteria after no treatment or oral administration of two different probiotic formula and their influence on the microbial ecosystem during and after the intervention and their association with the development of NEC. Weekly fecal samples were profiled by 16S rRNA sequencing and monitored for the presence of the probiotic bacteria by quantitative PCR. RESULTS Microbiota profiles differed significantly between the control group and both probiotic groups. Probiotic supplementation was associated with lower temporal variation as well as higher relative abundance of Bifidobacterium and Enterobacter combined with reduced abundance of Escherichia, Enterococcus, and Klebsiella. Colonization by probiotic bifidobacteria was observed in approximately 50% of infants although it remained transient in the majority of cases. A significantly reduced monthly incidence of NEC was observed in neonates supplemented with probiotics. CONCLUSION Our results demonstrate successful transient colonization by probiotic bacteria and a significant influence on the endogenous microbiota with a reduced abundance of bacterial taxa associated with the development of NEC. These results emphasize that probiotic supplementation may allow targeted manipulation of the enteric microbiota and confer a clinical benefit. (Clinical Trial Registry accession number: DRKS/GCTR 00021034).
Collapse
Affiliation(s)
- Niels van Best
- Institute of Medical Microbiology, RWTH University Hospital Aachen, RWTH University Aachen, Aachen, Germany,Department of Medical Microbiology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
| | | | - Paul Savelkoul
- Department of Medical Microbiology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
| | | | - Mathias W. Hornef
- Institute of Medical Microbiology, RWTH University Hospital Aachen, RWTH University Aachen, Aachen, Germany
| | - John Penders
- Department of Medical Microbiology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands,School of Public Health and Primary Care, Maastricht University, Maastricht, The Netherlands,CONTACT John Penders Department of Medical Microbiology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, HX6229, Maastricht, The Netherlands; Mathias Hornef Institute for Medical Microbiology; RWTH University Hospital; Pauwelsstr. 30, Aachen D-52074, Germany; Thorsten Orlikoswsky, Section of Neonatology, University Children’s Hospital, Pauwelsstr. 30, Aachen 52074, Germany
| |
Collapse
|
42
|
Lee JKF, Hern Tan LT, Ramadas A, Ab Mutalib NS, Lee LH. Exploring the Role of Gut Bacteria in Health and Disease in Preterm Neonates. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E6963. [PMID: 32977611 PMCID: PMC7579082 DOI: 10.3390/ijerph17196963] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/06/2020] [Accepted: 09/19/2020] [Indexed: 02/07/2023]
Abstract
The mortality rate of very preterm infants with birth weight <1500 g is as high as 15%. The survivors till discharge have a high incidence of significant morbidity, which includes necrotising enterocolitis (NEC), early-onset neonatal sepsis (EONS) and late-onset neonatal sepsis (LONS). More than 25% of preterm births are associated with microbial invasion of amniotic cavity. The preterm gut microbiome subsequently undergoes an early disruption before achieving bacterial maturation. It is postulated that bacterial gut colonisation at birth and postnatal intestinal dysbacteriosis precede the development of NEC and LONS in very preterm infants. In fact, bacterial colonization patterns in preterm infants greatly differ from term infants due to maternal chorioamnionitis, gestational age, delivery method, feeding type, antibiotic exposure and the environment factor in neonatal intensive care unit (NICU). In this regard, this review provides an overview on the gut bacteria in preterm neonates' meconium and stool. More than 50% of preterm meconium contains bacteria and the proportion increases with lower gestational age. Researchers revealed that the gut bacterial diversity is reduced in preterm infants at risk for LONS and NEC. Nevertheless, the association between gut dysbacteriosis and NEC is inconclusive with regards to relative bacteria abundance and between-sample beta diversity indices. With most studies show a disruption of the Proteobacteria and Firmicutes preceding the NEC. Hence, this review sheds light on whether gut bacteria at birth either alone or in combination with postnatal gut dysbacteriosis are associated with mortality and the morbidity of LONS and NEC in very preterm infants.
Collapse
Affiliation(s)
- Jimmy Kok-Foo Lee
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia; (J.K.-F.L.); (L.T.H.T.); (A.R.)
- Clinical School Johor Bahru, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Johor Bahru 80100, Malaysia
| | - Loh Teng Hern Tan
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia; (J.K.-F.L.); (L.T.H.T.); (A.R.)
| | - Amutha Ramadas
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia; (J.K.-F.L.); (L.T.H.T.); (A.R.)
| | - Nurul-Syakima Ab Mutalib
- UKM Medical Molecular Biology Institute (UMBI), UKM Medical Centre, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Learn-Han Lee
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia; (J.K.-F.L.); (L.T.H.T.); (A.R.)
| |
Collapse
|
43
|
Yu Y, Lu J, Oliphant K, Gupta N, Claud K, Lu L. Maternal administration of probiotics promotes gut development in mouse offsprings. PLoS One 2020; 15:e0237182. [PMID: 32764797 PMCID: PMC7413491 DOI: 10.1371/journal.pone.0237182] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/21/2020] [Indexed: 12/22/2022] Open
Abstract
Necrotizing enterocolitis is the most common gastrointestinal disorder in premature neonates. This disease is characterized by massive epithelial necrosis, gut barrier dysfunction and improper mucosal defense development. Studies have shown that probiotic administration can decrease NEC incidence and mortality. The proposed mechanisms of probiotics for the prevention of NEC are: promotion of intestinal development; improved barrier function through decreased apoptosis and improved mucin production; decreased expression of proinflammatory cytokines IL6, IL8, and TNFα, and modulation of microbiota dysbiosis in preterm infants. However, reported sepsis in the immunocompromised preterm host has deterred routine prophylactic administration of probiotics in the neonatal intensive care unit. We hypothesize that maternal administration of probiotics to pregnant mouse dams can recapitulate the beneficial effects observed in neonates fed with probiotics directly. We exposed pregnant mice to the probiotics and monitored the changes in the developing intestines of the offspring. Pregnant mice were fed daily with the probiotics Lactobacillus acidophilus and Bifidobacterium infantis (LB) from embryonic day15 to 2-week-old postnatally. Intraperitoneal administration of IL-1β in the pups was used to model proinflammatory insults. Sera were collected at 2 weeks of age and evaluated for inflammatory cytokines by enzyme-linked-immunosorbent-assay and gut permeability by Fluorescein isothiocyanate-dextran tracer assay. Ileal tissues were collected for the evaluation of apoptosis and proliferation of the intestinal epithelium; as well as mucin and tight junction integrity at mucosal surface by immunofluorescent staining. We find that maternal LB exposure facilitated intestinal epithelial cell differentiation, prevented loss of mucin and preserved the intestinal integrity and barrier function and decreased serum levels of IL-1β, TNF-α and IL-6 in the preweaned offsprings. in LB exposed pups. We demonstrate that maternal probiotic supplementation promotes gut maturation in developing offspring. This is potentially a safe alternative therapy to induce intestinal maturation and prevent prematurity-associated neonatal disorders.
Collapse
Affiliation(s)
- Yueyue Yu
- Department of Pediatrics, Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States of America
| | - Jing Lu
- Department of Pediatrics, Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States of America
| | - Kaitlyn Oliphant
- Department of Pediatrics, Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States of America
| | - Nikhilesh Gupta
- Department of Pediatrics, Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States of America
| | - Katerina Claud
- Department of Pediatrics, Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States of America
| | - Lei Lu
- Department of Pediatrics, Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
44
|
Rasmussen TS, Koefoed AK, Jakobsen RR, Deng L, Castro-Mejía JL, Brunse A, Neve H, Vogensen FK, Nielsen DS. Bacteriophage-mediated manipulation of the gut microbiome – promises and presents limitations. FEMS Microbiol Rev 2020; 44:507-521. [DOI: 10.1093/femsre/fuaa020] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 06/02/2020] [Indexed: 12/13/2022] Open
Abstract
ABSTRACT
Gut microbiome (GM) composition and function are linked to human health and disease, and routes for manipulating the GM have become an area of intense research. Due to its high treatment efficacy, the use of fecal microbiota transplantation (FMT) is generally accepted as a promising experimental treatment for patients suffering from GM imbalances (dysbiosis), e.g. caused by recurrent Clostridioides difficile infections (rCDI). Mounting evidence suggests that bacteriophages (phages) play a key role in successful FMT treatment by restoring the dysbiotic bacterial GM. As a refinement to FMT, removing the bacterial component of donor feces by sterile filtration, also referred to as fecal virome transplantation (FVT), decreases the risk of invasive infections caused by bacteria. However, eukaryotic viruses and prophage-encoded virulence factors remain a safety issue. Recent in vivo studies show how cascading effects are initiated when phage communities are transferred to the gut by e.g. FVT, which leads to changes in the GM composition, host metabolome, and improve host health such as alleviating symptoms of obesity and type-2-diabetes (T2D). In this review, we discuss the promises and limitations of FVT along with the perspectives of using FVT to treat various diseases associated with GM dysbiosis.
Collapse
Affiliation(s)
- Torben Sølbeck Rasmussen
- Section of Microbiology and Fermentation, Department of Food Science, University of Copenhagen, Rolighedsvej 26 4th floor - 1958, Frederiksberg, Denmark
| | - Anna Kirstine Koefoed
- Section of Microbiology and Fermentation, Department of Food Science, University of Copenhagen, Rolighedsvej 26 4th floor - 1958, Frederiksberg, Denmark
| | - Rasmus Riemer Jakobsen
- Section of Microbiology and Fermentation, Department of Food Science, University of Copenhagen, Rolighedsvej 26 4th floor - 1958, Frederiksberg, Denmark
| | - Ling Deng
- Section of Microbiology and Fermentation, Department of Food Science, University of Copenhagen, Rolighedsvej 26 4th floor - 1958, Frederiksberg, Denmark
| | - Josué L Castro-Mejía
- Section of Microbiology and Fermentation, Department of Food Science, University of Copenhagen, Rolighedsvej 26 4th floor - 1958, Frederiksberg, Denmark
| | - Anders Brunse
- Section of Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Ridebanevej 9, 2nd floor - 1870, Frederiksberg, Denmark
| | - Horst Neve
- Institute of Microbiology and Biotechnology, Max Rubner-Institut, Hermann-Weigmann-Straße 1 - 24103, Kiel, Germany
| | - Finn Kvist Vogensen
- Section of Microbiology and Fermentation, Department of Food Science, University of Copenhagen, Rolighedsvej 26 4th floor - 1958, Frederiksberg, Denmark
| | - Dennis Sandris Nielsen
- Section of Microbiology and Fermentation, Department of Food Science, University of Copenhagen, Rolighedsvej 26 4th floor - 1958, Frederiksberg, Denmark
| |
Collapse
|
45
|
Yang J, Yao S, Cheng K, Xu L, Hou L, Wei Y, Feng H, Yu X, Zhang X, Tong X, Li Z, Zhao Y. Comparison of Meconium Microbiome in Dizygotic and Monozygotic Twins Born by Caesarean Section (CS). Front Microbiol 2020; 11:1139. [PMID: 32582089 PMCID: PMC7283445 DOI: 10.3389/fmicb.2020.01139] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/05/2020] [Indexed: 01/27/2023] Open
Abstract
The early-life microbiota triggers life-long effects on physiological functions and health disorders. Previous studies in adult twins or animal models have revealed associations between host genetics and the harmonious microbiota. However, such associations may be obscured by the fact that each intra-pair of twins will continually encounter various environmental factors as they grow up. Here, we collected the meconium samples from nineteen dizygotic pairs (DZ, n = 38) and nine monozygotic pairs (MZ, n = 18) with cesarean delivery, and 16S rRNA gene sequencing was performed to profile the microbiome at birth. Diversity analysis showed that alpha diversity was not significantly different between two groups, whereas beta diversity of MZ twins was significantly lower than that of either DZ twins or unrelated individuals (i.e., randomly selected individual pairs of non-twinship) (p < 0.05). Two groups had very similar microbial classifications but different relative abundances of certain taxa including more Firmicutes (p = 0.05, Wilcoxon test) at the phylum level and lower abundances of five genera (p < 0.05) in DZ group compared to MZ group, including Rheinheimera, Proteus, SMB53, Sphingobium, and Megamonas. Co-occurrence analysis in each group showed slightly more complicated microbial interactions in DZ than MZ twins, although 22 shared bacterial genera co-existed in two groups, with both Rheinheimera and Megamonas having different centralities in their respective co-occurrence networks. Mean intra-class correlation coefficient (ICC) were also significantly higher for MZ (0.312) compared to DZ twins (0.138) (p < 0.05). The predicted microbial gene functions related to carbohydrate were higher in DZ group, whereas folding, sorting, degradation, cell motility pathways and energy metabolism were markedly over-represented in the microbiota of MZ group. In summary, our study uncovered that microbial diversity and components of the meconium microbiome between DZ and MZ twins were partially consistent with that in singleton neonates by cesarean delivery, but several distinctions related to the heritability supported genetic contributions to intestinal microbiome in early life.
Collapse
Affiliation(s)
- Jing Yang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Su Yao
- China Center of Industrial Culture Collection (CICC), China National Research Institute of Food & Fermentation Industries Co., Ltd., Beijing, China
| | - Kun Cheng
- China Center of Industrial Culture Collection (CICC), China National Research Institute of Food & Fermentation Industries Co., Ltd., Beijing, China
| | - Lili Xu
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| | - Lingling Hou
- Computational Genomics Lab, Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| | - Yuan Wei
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Huijun Feng
- China Center of Industrial Culture Collection (CICC), China National Research Institute of Food & Fermentation Industries Co., Ltd., Beijing, China
| | - Xuejian Yu
- China Center of Industrial Culture Collection (CICC), China National Research Institute of Food & Fermentation Industries Co., Ltd., Beijing, China
| | - Xin Zhang
- China Center of Industrial Culture Collection (CICC), China National Research Institute of Food & Fermentation Industries Co., Ltd., Beijing, China
| | - Xiaomei Tong
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| | - Zailing Li
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| | - Yangyu Zhao
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
46
|
Chen Y, Brook TC, Soe CZ, O'Neill I, Alcon-Giner C, Leelastwattanagul O, Phillips S, Caim S, Clarke P, Hall LJ, Hoyles L. Preterm infants harbour diverse Klebsiella populations, including atypical species that encode and produce an array of antimicrobial resistance- and virulence-associated factors. Microb Genom 2020; 6:e000377. [PMID: 32436839 PMCID: PMC7371107 DOI: 10.1099/mgen.0.000377] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 04/26/2020] [Indexed: 01/08/2023] Open
Abstract
Klebsiella spp. are frequently enriched in the gut microbiota of preterm neonates, and overgrowth is associated with necrotizing enterocolitis (NEC), nosocomial infections and late-onset sepsis. Little is known about the genomic and phenotypic characteristics of preterm-associated Klebsiella, as previous studies have focused on the recovery of antimicrobial-resistant isolates or culture-independent molecular analyses. The aim of this study was to better characterize preterm-associated Klebsiella populations using phenotypic and genotypic approaches. Faecal samples from a UK cohort of healthy and sick preterm neonates (n=109) were screened on MacConkey agar to isolate lactose-positive Enterobacteriaceae. Whole-genome sequences were generated for Klebsiella spp., and virulence and antimicrobial resistance genes identified. Antibiotic susceptibility profiling and in vitro macrophage and iron assays were undertaken for the Klebsiella strains. Metapangenome analyses with a manually curated genome dataset were undertaken to examine the diversity of Klebsiella oxytoca and related bacteria in a publicly available shotgun metagenome dataset. Approximately one-tenth of faecal samples harboured Klebsiella spp. (Klebsiella pneumoniae, 7.3 %; Klebsiella quasipneumoniae, 0.9 %; Klebsiella grimontii, 2.8 %; Klebsiella michiganensis, 1.8 %). Isolates recovered from NEC- and sepsis-affected infants and those showing no signs of clinical infection (i.e. 'healthy') encoded multiple β-lactamases. No difference was observed between isolates recovered from healthy and sick infants with respect to in vitro siderophore production (all encoded enterobactin in their genomes). All K. pneumoniae, K. quasipneumoniae, K. grimontii and K. michiganensis faecal isolates tested were able to reside and persist in macrophages, indicating their immune evasion abilities. Metapangenome analyses of published metagenomic data confirmed our findings regarding the presence of K. michiganensis in the preterm gut. There is little difference in the phenotypic and genomic characteristics of Klebsiella isolates recovered from healthy and sick infants. Identification of β-lactamases in all isolates may prove problematic when defining treatment regimens for NEC or sepsis, and suggests that healthy preterm infants contribute to the resistome. Refined analyses with curated sequence databases are required when studying closely related species present in metagenomic data.
Collapse
Affiliation(s)
- Yuhao Chen
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - Thomas C. Brook
- Department of Biomedical Sciences, Faculty of Science and Technology, University of Westminster, London, UK
| | - Cho Zin Soe
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Ian O'Neill
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Cristina Alcon-Giner
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Onnicha Leelastwattanagul
- Bioinformatics and Systems Biology Program, School of Bioresources and Technology, King Mongkut's University of Technology Thonburi (Bang Khun Thian Campus), Bangkok, Thailand
| | - Sarah Phillips
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Shabhonam Caim
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Paul Clarke
- Neonatal Intensive Care Unit, Norfolk and Norwich University Hospitals NHS Foundation Trust, Norwich, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Lindsay J. Hall
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Lesley Hoyles
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| |
Collapse
|
47
|
Lu J, Lu L, Yu Y, Baranowski J, Claud EC. Maternal administration of probiotics promotes brain development and protects offspring's brain from postnatal inflammatory insults in C57/BL6J mice. Sci Rep 2020; 10:8178. [PMID: 32424168 PMCID: PMC7235088 DOI: 10.1038/s41598-020-65180-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 04/29/2020] [Indexed: 12/12/2022] Open
Abstract
Neonatal morbidities are associated with long term neurological deficits in life and have also been associated with dysbiosis. We tested whether optimizing the neonate's microbiome through maternal probiotic supplementation can improve offspring's neurodevelopmental outcomes. Maternal LB supplementation, carried out by giving Lactobacillus acidophilus and Bifidobacterium infantis (LB) to pregnant C57/BL6J mice daily from E16 to weaning, significantly suppressed postnatal peripheral proinflammatory insult-induced systemic inflammation and normalized compromised blood-brain barrier permeability and tight junction protein expression in the offspring at pre-weaned age. Maternal LB exposure also regulated markers associated with leukocyte transendothelial migration, extracellular matrix injury and neuroinflammation. The suppressed neuroinflammation by maternal LB supplementation was associated with reduced astrocyte/microglia activation and downregulation of the transcriptional regulators CEBPD and IκBα. Furthermore, maternal LB supplementation promoted neuronal and oligodendrocyte progenitor cell development. Our study demonstrates the efficacy of maternal LB supplementation in modulating systemic and central nervous system inflammation as well as promoting neural/oligodendrocyte progenitor development in the offspring. This evidence suggests that maternal probiotic supplementation may be a safe and effective strategy to improve neurological outcomes in the offspring.
Collapse
Affiliation(s)
- Jing Lu
- The University of Chicago, Pritzker School of Medicine, Department of Pediatrics, Chicago, IL, 60637, USA
| | - Lei Lu
- The University of Chicago, Pritzker School of Medicine, Department of Pediatrics, Chicago, IL, 60637, USA
| | - Yueyue Yu
- The University of Chicago, Pritzker School of Medicine, Department of Pediatrics, Chicago, IL, 60637, USA
| | - Jillian Baranowski
- The University of Chicago, Pritzker School of Medicine, Chicago, IL, 60637, USA
| | - Erika C Claud
- The University of Chicago, Pritzker School of Medicine, Department of Pediatrics, Chicago, IL, 60637, USA.
| |
Collapse
|
48
|
Feeding Formula Eliminates the Necessity of Bacterial Dysbiosis and Induces Inflammation and Injury in the Paneth Cell Disruption Murine NEC Model in an Osmolality-Dependent Manner. Nutrients 2020; 12:nu12040900. [PMID: 32224880 PMCID: PMC7230818 DOI: 10.3390/nu12040900] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/19/2020] [Accepted: 03/24/2020] [Indexed: 01/08/2023] Open
Abstract
Necrotizing enterocolitis (NEC) remains a significant cause of morbidity and mortality in preterm infants. Formula feeding is a risk factor for NEC and osmolality, which is increased by the fortification that is required for adequate growth of the infant, has been suggested as a potential cause. Our laboratory has shown that Paneth cell disruption followed by induction of dysbiosis can induce NEC-like pathology in the absence of feeds. We hypothesized adding formula feeds to the model would exacerbate intestinal injury and inflammation in an osmolality-dependent manner. NEC-like injury was induced in 14-16 day-old C57Bl/6J mice by Paneth cell disruption with dithizone or diphtheria toxin, followed by feeding rodent milk substitute with varying osmolality (250-1491 mOsm/kg H2O). Animal weight, serum cytokines and osmolality, small intestinal injury, and cecal microbial composition were quantified. Paneth cell-disrupted mice fed formula had significant NEC scores compared to controls and no longer required induction of bacterial dysbiosis. Significant increases in serum inflammatory markers, small intestinal damage, and overall mortality were osmolality-dependent and not related to microbial changes. Overall, formula feeding in combination with Paneth cell disruption induced NEC-like injury in an osmolality-dependent manner, emphasizing the importance of vigilance in designing preterm infant feeds.
Collapse
|
49
|
Abstract
Necrotizing enterocolitis (NEC) is a poorly defined disease that primarily affects preterm infants. There has not been much progress in the prevention or treatment of NEC since it became recognized as a common problem in preterm infants. Reasons for this lack of progress include the likelihood that different diseases are being put under the same moniker of "NEC," similar to using "diabetes" for the different diseases it represents. In order to make progress, better delineation of the phenotypes that present as NEC will be necessary to clearly establish their pathophysiology, find specific and sensitive biomarkers, and establish preventative regimens. In this review, we summarize some of the entities that are being called NEC, discuss the pathophysiology of the most classic form of NEC, and provide an overview of how we might proceed in the future to make progress in this field.
Collapse
Affiliation(s)
- Josef Neu
- University of Florida, Gainesville, USA.
| |
Collapse
|
50
|
Elevated Coefficient of Variation in Total Fecal Bile Acids Precedes Diagnosis of Necrotizing Enterocolitis. Sci Rep 2020; 10:249. [PMID: 31937876 PMCID: PMC6959237 DOI: 10.1038/s41598-019-57178-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 12/24/2019] [Indexed: 12/15/2022] Open
Abstract
Accumulation of bile acids (BAs) may mediate development of necrotizing enterocolitis (NEC). Serial fecal samples were collected from premature infants with birth weight (BW) ≤ 1800 g, estimated gestational age (EGA) ≤ 32 weeks, and <30 days old prior to initiation of enteral feeding. Nine infants that developed Bell’s Stage ≥ II NEC were matched with control infants based on BW, EGA, day of life (DOL) enteral feeding was initiated and DOL of the first sample. From each subject, five samples matched by DOL collected were analyzed for BA levels and composition. Fifteen individual BA species were measured via LC-MS/MS and total BA levels were measured using the Diazyme Total Bile Acid Assay kit. No statistically significant differences in composition were observed between control and NEC at the level of individual species (p = 0.1133) or grouped BAs (p = 0.0742). However, there was a statistically significant difference (p = 0.000012) in the mean coefficient of variation (CV) between the two groups with infants developing NEC having more than four-fold higher mean CV than controls. Importantly, these variations occurred prior to NEC diagnosis. These data suggest fluctuations in total fecal BA levels could provide the basis for the first predictive clinical test for NEC.
Collapse
|