401
|
Effect of fecal microbiota transplantation route of administration on gut colonization and host response in preterm pigs. ISME JOURNAL 2018; 13:720-733. [PMID: 30367124 DOI: 10.1038/s41396-018-0301-z] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 09/26/2018] [Accepted: 10/04/2018] [Indexed: 02/06/2023]
Abstract
This study examined gut colonization patterns and host responses to fecal microbiota transplantation (FMT) by different administration routes after preterm birth. In two separate experiments, cesarean-delivered, preterm pigs were administered combined oral + rectal, or exclusively rectal donor feces, and compared with saline controls. After 5 days, stomach and colon bacterial compositions were determined by 16S rRNA gene amplicon sequencing, and organic acid metabolites measured. Further, gut pathology, mucosa bacterial adherence, and goblet cell density were assessed. FMT increased the relative abundance of obligate anaerobes in the colon without affecting total bacterial load. Bacteroides colonized recipients despite low abundance in the donor feces, whereas highly abundant Prevotella and Ruminococcaceae did not. Further, FMT changed carbohydrate metabolism from lactate to propionate production thereby increasing colonic pH. Besides, FMT preserved goblet cell mucin stores and reduced necrotizing enterocolitis incidence. Only rectal FMT increased the stomach-to-colon pH gradient and resistance to mucosa bacterial adhesion. Conversely, oral + rectal FMT increased bacterial adhesion, internal organ colonization, and overall mortality. Our results uncovered distinctions in bacterial colonization patterns along the gastrointestinal tract, as well as host tolerability between oral and rectal FMT administration in preterm newborns. Besides, FMT showed the potential to prevent necrotizing enterocolitis.
Collapse
|
402
|
Robertson RC, Manges AR, Finlay BB, Prendergast AJ. The Human Microbiome and Child Growth - First 1000 Days and Beyond. Trends Microbiol 2018; 27:131-147. [PMID: 30529020 DOI: 10.1016/j.tim.2018.09.008] [Citation(s) in RCA: 420] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 09/04/2018] [Accepted: 09/25/2018] [Indexed: 02/07/2023]
Abstract
The assembly of microbial communities within the gastrointestinal tract during early life plays a critical role in immune, endocrine, metabolic, and other host developmental pathways. Environmental insults during this period, such as food insecurity and infections, can disrupt this optimal microbial succession, which may contribute to lifelong and intergenerational deficits in growth and development. Here, we review the human microbiome in the first 1000 days - referring to the period from conception to 2 years of age - and using a developmental model, we examine the role of early microbial succession in growth and development. We propose that an 'undernourished' microbiome is intergenerational, thereby perpetuating growth impairments into successive generations. We also identify and discuss the intertwining host-microbe-environment interactions occurring prenatally and during early infancy, which may impair the trajectories of healthy growth and development, and explore their potential as novel microbial targets for intervention.
Collapse
Affiliation(s)
- Ruairi C Robertson
- Centre for Genomics & Child Health, Blizard Institute, Queen Mary University of London, UK.
| | - Amee R Manges
- University of British Columbia, School of Population and Public Health, Vancouver, BC, Canada; British Columbia Centre for Disease Control, Vancouver, BC, Canada
| | - B Brett Finlay
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - Andrew J Prendergast
- Centre for Genomics & Child Health, Blizard Institute, Queen Mary University of London, UK; Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| |
Collapse
|
403
|
Wejryd E, Martí M, Marchini G, Werme A, Jonsson B, Landberg E, Abrahamsson TR. Low Diversity of Human Milk Oligosaccharides is Associated with Necrotising Enterocolitis in Extremely Low Birth Weight Infants. Nutrients 2018; 10:nu10101556. [PMID: 30347801 PMCID: PMC6213888 DOI: 10.3390/nu10101556] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 10/12/2018] [Accepted: 10/12/2018] [Indexed: 12/27/2022] Open
Abstract
Difference in human milk oligosaccharides (HMO) composition in breast milk may be one explanation why some preterm infants develop necrotizing enterocolitis (NEC) despite being fed exclusively with breast milk. The aim of this study was to measure the concentration of 15 dominant HMOs in breast milk during the neonatal period and investigate how their levels correlated to NEC, sepsis, and growth in extremely low birth weight (ELBW; <1000 g) infants who were exclusively fed with breast milk. Milk was collected from 91 mothers to 106 infants at 14 and 28 days and at postmenstrual week 36. The HMOs were analysed with high-performance anion-exchange chromatography with pulsed amperometric detection. The HMOs diversity and the levels of Lacto-N-difucohexaose I were lower in samples from mothers to NEC cases, as compared to non-NEC cases at all sampling time points. Lacto-N-difucohexaose I is only produced by secretor and Lewis positive mothers. There were also significant but inconsistent associations between 3′-sialyllactose and 6′-sialyllactose and culture-proven sepsis and significant, but weak correlations between several HMOs and growth rate. Our results suggest that the variation in HMO composition in breast milk may be an important factor explaining why exclusively breast milk fed ELBW infants develop NEC.
Collapse
Affiliation(s)
- Erik Wejryd
- Department of Clinical and Experimental Medicine, Linköping University, 58183 Linköping, Sweden.
| | - Magalí Martí
- Department of Clinical and Experimental Medicine, Linköping University, 58183 Linköping, Sweden.
| | - Giovanna Marchini
- Department of Neonatology, Karolinska University Hospital, 17176 Stockholm, Sweden.
- Department of Women´s and Children´s Health, Karolinska Insitute, 17177 Stockholm, Sweden.
| | - Anna Werme
- Department of Clinical and Experimental Medicine, Linköping University, 58183 Linköping, Sweden.
| | - Baldvin Jonsson
- Department of Neonatology, Karolinska University Hospital, 17176 Stockholm, Sweden.
- Department of Women´s and Children´s Health, Karolinska Insitute, 17177 Stockholm, Sweden.
| | - Eva Landberg
- Department of Clinical and Experimental Medicine, Linköping University, 58183 Linköping, Sweden.
- Department of Clinical Chemistry, Linköping University, 58185 Linköping, Sweden.
| | - Thomas R Abrahamsson
- Department of Clinical and Experimental Medicine, Linköping University, 58183 Linköping, Sweden.
- Department of Pediatrics, Linköping University, 58183 Linköping, Sweden.
| |
Collapse
|
404
|
Pickard JM, Zeng MY, Caruso R, Núñez G. Gut microbiota: Role in pathogen colonization, immune responses, and inflammatory disease. Immunol Rev 2018; 279:70-89. [PMID: 28856738 DOI: 10.1111/imr.12567] [Citation(s) in RCA: 929] [Impact Index Per Article: 154.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The intestinal tract of mammals is colonized by a large number of microorganisms including trillions of bacteria that are referred to collectively as the gut microbiota. These indigenous microorganisms have co-evolved with the host in a symbiotic relationship. In addition to metabolic benefits, symbiotic bacteria provide the host with several functions that promote immune homeostasis, immune responses, and protection against pathogen colonization. The ability of symbiotic bacteria to inhibit pathogen colonization is mediated via several mechanisms including direct killing, competition for limited nutrients, and enhancement of immune responses. Pathogens have evolved strategies to promote their replication in the presence of the gut microbiota. Perturbation of the gut microbiota structure by environmental and genetic factors increases the risk of pathogen infection, promotes the overgrowth of harmful pathobionts, and the development of inflammatory disease. Understanding the interaction of the microbiota with pathogens and the immune system will provide critical insight into the pathogenesis of disease and the development of strategies to prevent and treat inflammatory disease.
Collapse
Affiliation(s)
- Joseph M Pickard
- Department of Pathology and Comprehensive Cancer Center, The University of Michigan Medical School, Ann Arbor, MI, USA
| | - Melody Y Zeng
- Department of Pathology and Comprehensive Cancer Center, The University of Michigan Medical School, Ann Arbor, MI, USA
| | - Roberta Caruso
- Department of Pathology and Comprehensive Cancer Center, The University of Michigan Medical School, Ann Arbor, MI, USA
| | - Gabriel Núñez
- Department of Pathology and Comprehensive Cancer Center, The University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
405
|
Bering SB. Human Milk Oligosaccharides to Prevent Gut Dysfunction and Necrotizing Enterocolitis in Preterm Neonates. Nutrients 2018; 10:nu10101461. [PMID: 30297668 PMCID: PMC6213229 DOI: 10.3390/nu10101461] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 10/04/2018] [Accepted: 10/05/2018] [Indexed: 12/25/2022] Open
Abstract
This review focuses on the evidence for health benefits of human milk oligosaccharides (HMOs) for preterm infants to stimulate gut adaptation and reduce the incidence of necrotizing enterocolitis (NEC) in early life. The health benefits of breastfeeding are partly explained by the abundant HMOs that serve as prebiotics and immunomodulators. Gut immaturity in preterm infants leads to difficulties in tolerating enteral feeding and bacterial colonization and a high sensitivity to NEC, particularly when breast milk is insufficient. Due to the immaturity of the preterm infants, their response to HMOs could be different from that in term infants. The concentration of HMOs in human milk is highly variable and there is no evidence to support a specifically adapted high concentration in preterm milk. Further, the gut microbiota is not only different but also highly variable after preterm birth. Studies in pigs as models for preterm infants indicate that HMO supplementation to formula does not mature the gut or prevent NEC during the first weeks after preterm birth and the effects may depend on a certain stage of gut maturity. Supplemented HMOs may become more important for gut protection in the preterm infants when the gut has reached a more mature phase.
Collapse
Affiliation(s)
- Stine Brandt Bering
- Comparative Paediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1958 Frederiksberg C, Denmark.
| |
Collapse
|
406
|
Staude B, Oehmke F, Lauer T, Behnke J, Göpel W, Schloter M, Schulz H, Krauss-Etschmann S, Ehrhardt H. The Microbiome and Preterm Birth: A Change in Paradigm with Profound Implications for Pathophysiologic Concepts and Novel Therapeutic Strategies. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7218187. [PMID: 30370305 PMCID: PMC6189679 DOI: 10.1155/2018/7218187] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 09/13/2018] [Indexed: 12/13/2022]
Abstract
Preterm birth poses a global challenge with a continuously increasing disease burden during the last decades. Advances in understanding the etiopathogenesis did not lead to a reduction of prematurely born infants so far. A balanced development of the host microbiome in early life is key for the maturation of the immune system and many other physiological functions. With the tremendous progress in new diagnostic possibilities, the contribution of microbiota changes to preterm birth and the acute and long-term sequelae of prematurity have come into the research focus. This review summarizes the latest advances in the understanding of microbiomes in the amniotic cavity and the female lower genital tract and how changes in microbiota structures contribute to preterm delivery. The exhibition of these highly vulnerable infants to the hostile environment in the neonatal intensive care unit necessarily entails the rapid colonization with a nonbalanced microbiome in a situation where the organism is still very prone and at an early stage of development. The global research efforts to decipher pathologic changes will pave the way to new pre- and postnatal therapeutic concepts.
Collapse
Affiliation(s)
- Birte Staude
- Department of General Pediatrics and Neonatology, Justus-Liebig-University and Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Feulgenstrasse 12, D-35392 Gießen, Germany
| | - Frank Oehmke
- Department of Gynecology and Obstetrics, Justus-Liebig-University, Feulgenstrasse 12, D-35392 Gießen, Germany
| | - Tina Lauer
- Department of General Pediatrics and Neonatology, Justus-Liebig-University and Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Feulgenstrasse 12, D-35392 Gießen, Germany
| | - Judith Behnke
- Department of General Pediatrics and Neonatology, Justus-Liebig-University and Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Feulgenstrasse 12, D-35392 Gießen, Germany
| | - Wolfgang Göpel
- Department of General Pediatrics, University Clinic of Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Michael Schloter
- Research Unit for Comparative Microbiome Analysis, Helmholtz Zentrum München GmbH, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| | - Holger Schulz
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research (DZL), Max-Lebsche-Platz 31, D-81377 Munich, Germany
| | - Susanne Krauss-Etschmann
- Research Center Borstel, Leibniz-Center for Medicine and Biosciences, Borstel, Germany, Member of the German Center for Lung Research (DZL), Germany
- Institute of Experimental Medicine, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Harald Ehrhardt
- Department of General Pediatrics and Neonatology, Justus-Liebig-University and Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Feulgenstrasse 12, D-35392 Gießen, Germany
| |
Collapse
|
407
|
Robinson JL, Smith VA, Stoll B, Agarwal U, Premkumar MH, Lau P, Cruz SM, Manjarin R, Olutoye O, Burrin DG, Marini JC. Prematurity reduces citrulline-arginine-nitric oxide production and precedes the onset of necrotizing enterocolitis in piglets. Am J Physiol Gastrointest Liver Physiol 2018; 315:G638-G649. [PMID: 30048597 PMCID: PMC6415715 DOI: 10.1152/ajpgi.00198.2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Necrotizing enterocolitis (NEC) is associated with low plasma arginine and vascular dysfunction. It is not clear whether low intestinal citrulline production, the precursor for arginine synthesis, occurs before and thus predisposes to NEC or if it results from tissue damage. This study was designed to test the hypothesis that whole body rates of citrulline, arginine, and nitric oxide synthesis are low in premature pigs and that they precede NEC. Piglets delivered by cesarean section at 103 days [preterm (PT)], 110 days [near-term (NT)], or 114 days [full-term (FT)] of gestation were given total parenteral nutrition and after 2 days orogastrically fed infant formula for 42 h to induce NEC. Citrulline and arginine fluxes were determined before and during the feeding protocol. Gross macroscopic and histological NEC scores and plasma fatty acid binding protein (iFABP) concentration were determined as indicators of NEC. Intestinal gene expression for enzymes of the arginine pathway were quantitated. A lower ( P < 0.05) survival rate was observed for PT (8/27) than for NT (9/9) and FT pigs (11/11). PT pigs had higher macroscopic gross ( P < 0.05) and histological NEC ( P < 0.05) scores and iFABP concentration ( P < 0.05) than pigs of more advanced gestational age. PT pigs had lower citrulline production and arginine fluxes ( P < 0.05) throughout and a reduced gene expression in genes of the citrulline-arginine pathway. In summary, intestinal enzyme expression and whole body citrulline and arginine fluxes were reduced in PT pigs compared with animals of more advance gestational age and preceded the development of NEC. NEW & NOTEWORTHY Arginine supplementation prevents necrotizing enterocolitis (NEC), the most common gastrointestinal emergency of prematurity. Citrulline (precursor for arginine) production is reduced during NEC, and this is believed to be a consequence of intestinal damage. In a swine model of NEC, we show that intestinal gene expression of the enzymes for citrulline production and whole body citrulline and arginine fluxes are reduced and precede the onset of NEC in premature pigs. Reduced citrulline production during prematurity may be a predisposition to NEC.
Collapse
Affiliation(s)
- Jason L. Robinson
- 1United States Department of Agriculture, Agricultural Research Service, Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Victoria A. Smith
- 2Department of Animal Science, California Polytechnic State University, San Luis Obispo, California
| | - Barbara Stoll
- 1United States Department of Agriculture, Agricultural Research Service, Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Umang Agarwal
- 1United States Department of Agriculture, Agricultural Research Service, Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Muralidhar H. Premkumar
- 3Division of Neonatology, Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas
| | - Patricio Lau
- 4Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas
| | - Stephanie M. Cruz
- 4Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas
| | - Rodrigo Manjarin
- 2Department of Animal Science, California Polytechnic State University, San Luis Obispo, California
| | - Oluyinka Olutoye
- 4Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas
| | - Douglas G. Burrin
- 1United States Department of Agriculture, Agricultural Research Service, Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Juan C. Marini
- 1United States Department of Agriculture, Agricultural Research Service, Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas,5Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
408
|
Calprotectin and necrotizing enterocolitis: it's not the assay, it's the definition. J Perinatol 2018; 38:1285-1286. [PMID: 30108341 DOI: 10.1038/s41372-018-0201-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 07/16/2018] [Indexed: 11/09/2022]
|
409
|
Hoggard M, Vesty A, Wong G, Montgomery JM, Fourie C, Douglas RG, Biswas K, Taylor MW. Characterizing the Human Mycobiota: A Comparison of Small Subunit rRNA, ITS1, ITS2, and Large Subunit rRNA Genomic Targets. Front Microbiol 2018; 9:2208. [PMID: 30283425 PMCID: PMC6157398 DOI: 10.3389/fmicb.2018.02208] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Accepted: 08/29/2018] [Indexed: 12/28/2022] Open
Abstract
Interest in the human microbiome has increased dramatically in the last decade. However, much of this research has focused on bacteria, while the composition and roles of their fungal counterparts remain less understood. Furthermore, a variety of methodological approaches have been applied, and the comparability between studies is unclear. This study compared four primer pairs targeting the small subunit (SSU) rRNA (18S), ITS1, ITS2, and large subunit (LSU) rRNA (26S) genomic regions for their ability to accurately characterize fungal communities typical of the human mycobiota. All four target regions of 21 individual fungal mock community taxa were capable of being amplified adequately and sequenced. Mixed mock community analyses revealed marked variability in the ability of each primer pair to accurately characterize a complex community. ITS target regions outperformed LSU and SSU. Of the ITS regions, ITS1 failed to generate sequences for Yarrowia lipolytica and all three Malassezia species when in a mixed community. These findings were further supported in studies of human sinonasal and mouse fecal samples. Based on these analyses, previous studies using ITS1, SSU, or LSU markers may omit key taxa that are identified by the ITS2 marker. Of methods commonly used in human mycobiota studies to date, we recommend selection of the ITS2 marker. Further investigation of more recently developed fungal primer options will be essential to ultimately determine the optimal methodological approach by which future human mycobiota studies ought to be standardized.
Collapse
Affiliation(s)
- Michael Hoggard
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| | - Anna Vesty
- School of Medicine, The University of Auckland, Auckland, New Zealand.,Microbiology Laboratory, LabPLUS, Auckland City Hospital, Auckland, New Zealand
| | - Giselle Wong
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| | - Johanna M Montgomery
- Department of Physiology, Centre for Brain Research, The University of Auckland, Auckland, New Zealand
| | - Chantelle Fourie
- Department of Physiology, Centre for Brain Research, The University of Auckland, Auckland, New Zealand
| | - Richard G Douglas
- School of Medicine, The University of Auckland, Auckland, New Zealand
| | - Kristi Biswas
- School of Medicine, The University of Auckland, Auckland, New Zealand
| | - Michael W Taylor
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
410
|
Ho TTB, Groer MW, Kane B, Yee AL, Torres BA, Gilbert JA, Maheshwari A. Dichotomous development of the gut microbiome in preterm infants. MICROBIOME 2018; 6:157. [PMID: 30208950 PMCID: PMC6136210 DOI: 10.1186/s40168-018-0547-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 09/03/2018] [Indexed: 05/31/2023]
Abstract
BACKGROUND Preterm infants are at risk of developing intestinal dysbiosis with an increased proportion of Gammaproteobacteria. In this study, we sought the clinical determinants of the relative abundance of feces-associated Gammaproteobacteria in very low birth weight (VLBW) infants. Fecal microbiome was characterized at ≤ 2 weeks and during the 3rd and 4th weeks after birth, by 16S rRNA amplicon sequencing. Maternal and infant clinical characteristics were extracted from electronic medical records. Data were analyzed by linear mixed modeling and linear regression. RESULTS Clinical data and fecal microbiome profiles of 45 VLBW infants (gestational age 27.9 ± 2.2 weeks; birth weight 1126 ± 208 g) were studied. Three stool samples were analyzed for each infant at mean postnatal ages of 9.9 ± 3, 20.7 ± 4.1, and 29.4 ± 4.9 days. The average relative abundance of Gammaproteobacteria was 42.5% (0-90%) at ≤ 2 weeks, 69.7% (29.9-86.9%) in the 3rd, and 75.5% (54.5-86%) in the 4th week (p < 0.001). Hierarchical and K-means clustering identified two distinct subgroups: cluster 1 started with comparatively low abundance that increased with time, whereas cluster 2 began with a greater abundance at ≤ 2 weeks (p < 0.001) that decreased over time. Both groups resembled each other by the 3rd week. Single variants of Klebsiella and Staphylococcus described variance in community structure between clusters and were shared between all infants, suggesting a common, hospital-derived source. Fecal Gammaproteobacteria was positively associated with vaginal delivery and antenatal steroids. CONCLUSIONS We detected a dichotomy in gut microbiome assembly in preterm infants: some preterm infants started with low relative gammaproteobacterial abundance in stool that increased as a function of postnatal age, whereas others began with and maintained high abundance. Vaginal birth and antenatal steroids were identified as predictors of Gammaproteobacteria abundance in the early (≤ 2 weeks) and later (3rd and 4th weeks) stool samples, respectively. These findings are important in understanding the development of the gut microbiome in premature infants.
Collapse
Affiliation(s)
- Thao T. B. Ho
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL USA
| | - Maureen W. Groer
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL USA
- College of Nursing, University of South Florida, Tampa, FL USA
| | - Bradley Kane
- College of Nursing, University of South Florida, Tampa, FL USA
| | - Alyson L. Yee
- Interdisciplinary Scientist Training Program, University of Chicago, Chicago, IL USA
- Microbiome Center, University of Chicago, Chicago, IL USA
- Department of Surgery, University of Chicago, Chicago, IL USA
| | - Benjamin A. Torres
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL USA
| | - Jack A. Gilbert
- Microbiome Center, University of Chicago, Chicago, IL USA
- Department of Surgery, University of Chicago, Chicago, IL USA
- Argonne National Laboratory, Chicago, IL USA
| | - Akhil Maheshwari
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL USA
- Department of Pediatrics, Johns Hopkins University, 1800 Orleans St, JHCC 8530, Baltimore, MD 21287 USA
| |
Collapse
|
411
|
Olson JK, Navarro JB, Allen JM, McCulloh CJ, Mashburn-Warren L, Wang Y, Varaljay VA, Bailey MT, Goodman SD, Besner GE. An enhanced Lactobacillus reuteri biofilm formulation that increases protection against experimental necrotizing enterocolitis. Am J Physiol Gastrointest Liver Physiol 2018; 315:G408-G419. [PMID: 29848024 PMCID: PMC6415713 DOI: 10.1152/ajpgi.00078.2018] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 05/09/2018] [Accepted: 05/23/2018] [Indexed: 02/06/2023]
Abstract
One significant drawback of current probiotic therapy for the prevention of necrotizing enterocolitis (NEC) is the need for at least daily administration because of poor probiotic persistence after enteral administration, increasing the risk of the probiotic bacteria causing bacteremia or sepsis if the intestines are already compromised. We previously showed that the effectiveness of Lactobacillus reuteri ( Lr) in preventing NEC is enhanced when Lr is grown as a biofilm on the surface of dextranomer microspheres (DM). Here we sought to test the efficacy of Lr administration by manipulating the Lr biofilm state with the addition of biofilm-promoting substances (sucrose and maltose) to DM or by mutating the Lr gtfW gene (encoding an enzyme central to biofilm production). Using an animal model of NEC, we determined that Lr adhered to sucrose- or maltose-loaded DM significantly reduced histologic injury, improved host survival, decreased intestinal permeability, reduced intestinal inflammation, and altered the gut microbiome compared with Lr adhered to unloaded DM. These effects were abolished when DM or GtfW were absent from the Lr inoculum. This demonstrates that a single dose of Lr in its biofilm state decreases NEC incidence. Importantly, preloading DM with sucrose or maltose further enhances Lr protection against NEC in a GtfW-dependent fashion, demonstrating the tunability of the approach and the potential to use other cargos to enhance future probiotic formulations. NEW & NOTEWORTHY Previous clinical trials of probiotics to prevent necrotizing enterocolitis have had variable results. In these studies, probiotics were delivered in their planktonic, free-living form. We have developed a novel probiotic delivery system in which Lactobacillus reuteri (Lr) is delivered in its biofilm state. In a model of experimental necrotizing enterocolitis, this formulation significantly reduces intestinal inflammation and permeability, improves survival, and preserves the natural gut microflora compared with the administration of Lr in its free-living form.
Collapse
Affiliation(s)
- Jacob K Olson
- Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, Department of Pediatric Surgery, Nationwide Children's Hospital , Columbus, Ohio
| | - Jason B Navarro
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital , Columbus, Ohio
| | - Jacob M Allen
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital , Columbus, Ohio
| | - Christopher J McCulloh
- Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, Department of Pediatric Surgery, Nationwide Children's Hospital , Columbus, Ohio
| | - Lauren Mashburn-Warren
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital , Columbus, Ohio
| | - Yijie Wang
- Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, Department of Pediatric Surgery, Nationwide Children's Hospital , Columbus, Ohio
| | - Vanessa A Varaljay
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital , Columbus, Ohio
| | - Michael T Bailey
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital , Columbus, Ohio
| | - Steven D Goodman
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital , Columbus, Ohio
| | - Gail E Besner
- Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, Department of Pediatric Surgery, Nationwide Children's Hospital , Columbus, Ohio
| |
Collapse
|
412
|
Ma YP, Ma JY, Tong XM. [A review of the relationship between gut microbiome and necrotizing enterocolitis in preterm infants]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2018; 20:680-685. [PMID: 30111480 PMCID: PMC7389748 DOI: 10.7499/j.issn.1008-8830.2018.08.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 07/03/2018] [Indexed: 06/08/2023]
Abstract
Necrotizing enterocolitis (NEC) is a common severe gastrointestinal disease in preterm infants. The morbidity and mortality of NEC are negatively correlated with the gestational age and birth weight. In addition to causing a variety of gastrointestinal complications, NEC can also cause neurodevelopmental impairment. Recently, many studies have found that gut microbiome dysbiosis plays an important part in the pathogenesis of NEC. It is helpful to explore the relationship between gut microbiome and NEC for the early diagnosis and severity prediction of NEC. Researchers have paid much attention to the role of probiotics in reducing the morbidity and mortality of NEC in preterm infants. It's controversial as to whether probiotics is effective and safe in clinical application. This article will review the relationship between the development of gut microbiome and NEC in preterm infants, as well as the preventive effect of probiotics on NEC.
Collapse
Affiliation(s)
- Yuan-Pei Ma
- Department of Pediatrics, Peking University Third Hospital, Beijing 100191, China.
| | | | | |
Collapse
|
413
|
Ma YP, Ma JY, Tong XM. [A review of the relationship between gut microbiome and necrotizing enterocolitis in preterm infants]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2018; 20:680-685. [PMID: 30111480 PMCID: PMC7389748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 07/03/2018] [Indexed: 08/01/2024]
Abstract
Necrotizing enterocolitis (NEC) is a common severe gastrointestinal disease in preterm infants. The morbidity and mortality of NEC are negatively correlated with the gestational age and birth weight. In addition to causing a variety of gastrointestinal complications, NEC can also cause neurodevelopmental impairment. Recently, many studies have found that gut microbiome dysbiosis plays an important part in the pathogenesis of NEC. It is helpful to explore the relationship between gut microbiome and NEC for the early diagnosis and severity prediction of NEC. Researchers have paid much attention to the role of probiotics in reducing the morbidity and mortality of NEC in preterm infants. It's controversial as to whether probiotics is effective and safe in clinical application. This article will review the relationship between the development of gut microbiome and NEC in preterm infants, as well as the preventive effect of probiotics on NEC.
Collapse
Affiliation(s)
- Yuan-Pei Ma
- Department of Pediatrics, Peking University Third Hospital, Beijing 100191, China.
| | | | | |
Collapse
|
414
|
Rusconi B, Jiang X, Sidhu R, Ory DS, Warner BB, Tarr PI. Gut Sphingolipid Composition as a Prelude to Necrotizing Enterocolitis. Sci Rep 2018; 8:10984. [PMID: 30030452 PMCID: PMC6054655 DOI: 10.1038/s41598-018-28862-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 06/22/2018] [Indexed: 12/21/2022] Open
Abstract
Necrotizing enterocolitis (NEC) remains a major challenge in neonatology. Little is known about NEC pathophysiology apart from the presence of pre-event gut dysbiosis. Here, we applied broad range metabolomics to stools obtained 1-5 days before NEC developed from 9 cases (9 samples) and 19 (32 samples) controls matched for gestational age at birth and birth weight. The 764 identified metabolites identified six pathways that differ between cases and controls. We pursued sphingolipid metabolism because cases had decreased ceramides and increased sphingomyelins compared to controls, and because of the relevance of sphingolipids to human inflammatory disorders. Targeted analysis of samples from 23 cases and 46 controls confirmed the initial broad range observations. While metabolites provided only 73% accuracy of classification by machine learning, hierarchical clustering defined a sphingolipid associated grouping that contained 60% of the cases but only 13% of the controls, possibly identifying a pathophysiologically distinct subset of NEC. The clustering did not associate with any of the analyzed clinical and sample variables. We conclude that there are significant changes in sphingolipid metabolism components in pre-NEC stools compared to controls, but our data urge circumspection before using sphingolipids as broadly applicable predictive biomarkers.
Collapse
Affiliation(s)
- B Rusconi
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - X Jiang
- Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - R Sidhu
- Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - D S Ory
- Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - B B Warner
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - P I Tarr
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.
- Department of Molecular Microbiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
415
|
Sun J, Li Y, Pan X, Nguyen DN, Brunse A, Bojesen AM, Rudloff S, Mortensen MS, Burrin DG, Sangild PT. Human Milk Fortification with Bovine Colostrum Is Superior to Formula-Based Fortifiers to Prevent Gut Dysfunction, Necrotizing Enterocolitis, and Systemic Infection in Preterm Pigs. JPEN J Parenter Enteral Nutr 2018; 43:252-262. [PMID: 29992630 DOI: 10.1002/jpen.1422] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 06/04/2018] [Accepted: 06/05/2018] [Indexed: 12/29/2022]
Abstract
BACKGROUND Fortification of donor human milk (DHM) is required for optimal growth of very preterm infants, but there are concerns of more gut dysfunction and necrotizing enterocolitis (NEC) when using formula-based fortifiers (FFs), especially soon after birth. Intact bovine colostrum (BC) is rich in nutrients and bioactive factors, and protects against NEC in preterm pigs. We hypothesized that fortification of DHM with BC is superior to FFs to prevent gut dysfunction and infections when provided shortly after preterm birth. METHODS Two FF products, Enfamil (ENF; intact protein, vegetable oil) and PreNAN+Nutrilon (NAN; extensively hydrolyzed protein, maltodextrin), were compared with BC as fortifier to DHM fed to preterm pigs for 5 days. RESULTS Relative to the DHM+BC group, DHM+FF groups had higher diarrhea score and lower hexose uptake and lactase activity, and specifically the DHM+NAN group showed higher gut permeability, NEC score, more mucosa-adherent bacteria with altered gut microbiota structure (ie, lower diversity, increased Enterococcus, decreased Staphylococcus abundance). Both DHM+FF groups showed higher expression of intestinal cytokine and inflammation-related genes, more gut-derived bacteria in the bone marrow, lower density of mucin-containing goblet cells, and slightly higher colon lactate, stomach pH and acetate, and blood neutrophil-to-lymphocyte levels than the DHM+BC group. CONCLUSIONS Used as a fortifier to DHM, BC is superior to FFs to support gut function, nutrient absorption, and bacterial defense mechanisms in preterm pigs. It is important to optimize the composition of nutrient fortifiers for preterm infants fed human milk.
Collapse
Affiliation(s)
- Jing Sun
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Yanqi Li
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Xiaoyu Pan
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Duc Ninh Nguyen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anders Brunse
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anders M Bojesen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Silvia Rudloff
- Institute of Nutritional Science, Justus-Liebig-University Giessen , Giessen, Germany
| | | | - Douglas G Burrin
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Per T Sangild
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Pediatrics and Adolescent Medicine, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
416
|
Probiotics for Preterm Infants: A Strain-Specific Systematic Review and Network Meta-analysis. J Pediatr Gastroenterol Nutr 2018; 67:103-122. [PMID: 29384838 DOI: 10.1097/mpg.0000000000001897] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
OBJECTIVES Several randomized controlled trials (RCTs) on the use of probiotics to reduce morbidity and mortality in preterm infants have provided inconsistent results. Although meta-analyses that group all of the used strains together are suggesting efficacy, it is not possible to determine the most effective strain that is more relevant to the clinician. We therefore used a network meta-analysis (NMA) approach to identify strains with greatest efficacy. METHODS A PubMed search identified placebo-controlled or head-to-head RCTs investigating probiotics in preterm infants. From trials that recorded mortality, necrotizing enterocolitis, late-onset sepsis, or time until full enteral feeding as outcomes, data were extracted and Bayesian hierarchical random-effects models were run to construct a NMA. RESULTS Fifty-one RCTs involving 11,231 preterm infants were included. Most strains or combinations of strains were only studied in one or a few RCTs. Only 3 of 25 studied probiotic treatment combinations showed significant reduction in mortality rates. Seven treatments reduced necrotizing enterocolitis incidence, 2 reduced late-onset sepsis, and 3 reduced time until full enteral feeding. There was no clear overlap of strains, which were effective on multiple outcome domains. CONCLUSIONS This NMA showed efficacy in reducing mortality and morbidity only in a minority of the studied strains or combinations. This may be due to an inadequate number, or size, of RCTs, or due to a true lack of effect for certain species. Further large and adequately powered RCTs using strains with the greatest apparent efficacy will be needed to more precisely define optimal treatment strategies.
Collapse
|
417
|
Hourigan SK, Subramanian P, Hasan NA, Ta A, Klein E, Chettout N, Huddleston K, Deopujari V, Levy S, Baveja R, Clemency NC, Baker RL, Niederhuber JE, Colwell RR. Comparison of Infant Gut and Skin Microbiota, Resistome and Virulome Between Neonatal Intensive Care Unit (NICU) Environments. Front Microbiol 2018; 9:1361. [PMID: 29988506 PMCID: PMC6026636 DOI: 10.3389/fmicb.2018.01361] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 06/05/2018] [Indexed: 01/08/2023] Open
Abstract
Background: There is a growing move to provide care for premature infants in a single family, private room neonatal intensive care unit (NICU) in place of the traditional shared space, open bay NICU. The resultant effect on the developing neonatal microbiota is unknown. Study Design: Stool and groin skin swabs were collected from infants in a shared-space NICU (old NICU) and a single-family room NICU (new NICU) on the same hospital campus. Metagenomic sequencing was performed and data analyzed by CosmosID bioinformatics software package. Results: There were no significant differences between the cohorts in gestational age, length of stay, and delivery mode; infants in the old NICU received significantly more antibiotics (p = 0.03). Differentially abundant antimicrobial resistance genes and virulence associated genes were found between the cohorts in stool and skin, with more differentially abundant antimicrobial resistance genes in the new NICU. The entire bacterial microbiota analyzed to the genus level significantly differed between cohorts in skin (p = 0.0001) but not in stool samples. There was no difference in alpha diversity between the two cohorts. DNA viruses and fungi were detected but did not differ between cohorts. Conclusion: Differences were seen in the resistome and virulome between the two cohorts with more differentially abundant antimicrobial resistance genes in the new NICU. This highlights the influence that different NICU environments can have on the neonatal microbiota. Whether the differences were due to the new NICU being a single-family NICU or located in a newly constructed building warrants exploration. Long term health outcomes from the differences observed must be followed longitudinally.
Collapse
Affiliation(s)
- Suchitra K Hourigan
- Department of Pediatrics, Inova Children's Hospital, Falls Church, VA, United States.,Inova Translational Medicine Institute, Falls Church, VA, United States.,Department of Pediatric Gastroenterology, Pediatric Specialists of Virginia, Fairfax, VA, United States
| | | | | | - Allison Ta
- Department of Pediatrics, Inova Children's Hospital, Falls Church, VA, United States
| | - Elisabeth Klein
- Inova Translational Medicine Institute, Falls Church, VA, United States
| | - Nassim Chettout
- Inova Translational Medicine Institute, Falls Church, VA, United States
| | - Kathi Huddleston
- Inova Translational Medicine Institute, Falls Church, VA, United States
| | - Varsha Deopujari
- Inova Translational Medicine Institute, Falls Church, VA, United States
| | - Shira Levy
- Inova Translational Medicine Institute, Falls Church, VA, United States
| | - Rajiv Baveja
- Fairfax Neonatal Associates PC, Falls Church, VA, United States
| | - Nicole C Clemency
- Inova Translational Medicine Institute, Falls Church, VA, United States
| | - Robin L Baker
- Fairfax Neonatal Associates PC, Falls Church, VA, United States
| | | | | |
Collapse
|
418
|
Call L, Stoll B, Oosterloo B, Ajami N, Sheikh F, Wittke A, Waworuntu R, Berg B, Petrosino J, Olutoye O, Burrin D. Metabolomic signatures distinguish the impact of formula carbohydrates on disease outcome in a preterm piglet model of NEC. MICROBIOME 2018; 6:111. [PMID: 29921329 PMCID: PMC6009052 DOI: 10.1186/s40168-018-0498-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 06/08/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND Major risk factors for necrotizing enterocolitis (NEC) include premature birth and formula feeding in the context of microbial colonization of the gastrointestinal tract. We previously showed that feeding formula composed of lactose vs. corn syrup solids protects against NEC in preterm pigs; however, the microbial and metabolic effects of these different carbohydrates used in infant formula has not been explored. OBJECTIVE Our objective was to characterize the effects of lactose- and corn syrup solid-based formulas on the metabolic and microbial profiles of preterm piglets and to determine whether unique metabolomic or microbiome signatures correlate with severity or incidence of NEC. DESIGN/METHODS Preterm piglets (103 days gestation) were given total parenteral nutrition (2 days) followed by gradual (5 days) advancement of enteral feeding of formulas matched in nutrient content but containing either lactose (LAC), corn syrup solids (CSS), or 1:1 mix (MIX). Gut contents and mucosal samples were collected and analyzed for microbial profiles by sequencing the V4 region of the 16S rRNA gene. Metabolomic profiles of cecal contents and plasma were analyzed by LC/GC mass spectrometry. RESULTS NEC incidence was 14, 50, and 44% in the LAC, MIX, and CSS groups, respectively. The dominant classes of bacteria were Bacilli, Clostridia, and Gammaproteobacteria. The number of observed OTUs was lowest in colon contents of CSS-fed pigs. CSS-based formula was associated with higher Bacilli and lower Clostridium from clusters XIVa and XI in the colon. NEC was associated with decreased Gammaproteobacteria in the stomach and increased Clostridium sensu stricto in the ileum. Plasma from NEC piglets was enriched with metabolites of purine metabolism, aromatic amino acid metabolism, and bile acids. Markers of glycolysis, e.g., lactate, were increased in the cecal contents of CSS-fed pigs and in plasma of pigs which developed NEC. CONCLUSIONS Feeding formula containing lactose is not completely protective against NEC, yet selects for greater microbial richness associated with changes in Bacilli and Clostridium and lower NEC incidence. We conclude that feeding preterm piglets a corn syrup solid vs. lactose-based formula increases the incidence of NEC and produces distinct metabolomic signatures despite modest changes in microbiome profiles.
Collapse
Affiliation(s)
- Lee Call
- Department Pediatric Gastroenterology, Hepatology, and Nutrition, USDA-ARS Children’s Nutrition Research Center, 1100 Bates Ave, Houston, TX 77030 USA
| | - Barbara Stoll
- Department Pediatric Gastroenterology, Hepatology, and Nutrition, USDA-ARS Children’s Nutrition Research Center, 1100 Bates Ave, Houston, TX 77030 USA
| | - Berthe Oosterloo
- Department Pediatric Gastroenterology, Hepatology, and Nutrition, USDA-ARS Children’s Nutrition Research Center, 1100 Bates Ave, Houston, TX 77030 USA
| | - Nadim Ajami
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, One Baylor Plaza, MS BCM385, Houston, TX 77030 USA
| | - Fariha Sheikh
- Division of Pediatric Surgery, Baylor College of Medicine, 6701 Fannin St, Suite 1210, Houston, TX 77030 USA
| | - Anja Wittke
- Mead Johnson Pediatric Nutrition Institute, 2400 W Lloyd Expressway, Evansville, IN 47712 USA
| | - Rosaline Waworuntu
- Mead Johnson Pediatric Nutrition Institute, 2400 W Lloyd Expressway, Evansville, IN 47712 USA
| | - Brian Berg
- Mead Johnson Pediatric Nutrition Institute, 2400 W Lloyd Expressway, Evansville, IN 47712 USA
| | - Joseph Petrosino
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, One Baylor Plaza, MS BCM385, Houston, TX 77030 USA
| | - Oluyinka Olutoye
- Division of Pediatric Surgery, Baylor College of Medicine, 6701 Fannin St, Suite 1210, Houston, TX 77030 USA
| | - Douglas Burrin
- Department Pediatric Gastroenterology, Hepatology, and Nutrition, USDA-ARS Children’s Nutrition Research Center, 1100 Bates Ave, Houston, TX 77030 USA
| |
Collapse
|
419
|
Abstract
PURPOSE OF REVIEW Necrotizing enterocolitis (NEC) is a devastating disease that predominately affects premature neonates. The pathogenesis of NEC is multifactorial and poorly understood. Risk factors include low birth weight, formula-feeding, hypoxic/ischemic insults, and microbial dysbiosis. This review focuses on our current understanding of the diagnosis, management, and pathogenesis of NEC. RECENT FINDINGS Recent findings identify specific mucosal cell types as potential therapeutic targets in NEC. Despite a broadly accepted view that bacterial colonization plays a key role in NEC, characteristics of bacterial populations associated with this disease remain elusive. The use of probiotics such as lactobacilli and bifidobacteria has been studied in numerous trials, but there is a lack of consensus regarding specific strains and dosing. Although growth factors found in breast milk such as epidermal growth factor and heparin-binding epidermal growth factor may be useful in disease prevention, developing new therapeutic interventions in NEC critically depends on better understanding of its pathogenesis. SUMMARY NEC is a leading cause of morbidity and mortality in premature neonates. Recent data confirm that growth factors and certain bacteria may offer protection against NEC. Further studies are needed to better understand the complex pathogenesis of NEC.
Collapse
|
420
|
Reed BD, Schibler KR, Deshmukh H, Ambalavanan N, Morrow AL. The Impact of Maternal Antibiotics on Neonatal Disease. J Pediatr 2018; 197:97-103.e3. [PMID: 29551319 PMCID: PMC6028045 DOI: 10.1016/j.jpeds.2018.01.056] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 01/17/2018] [Accepted: 01/19/2018] [Indexed: 01/13/2023]
Abstract
OBJECTIVES We examined the impact of prenatal exposure to maternal antibiotics on risk of necrotizing enterocolitis (NEC), late onset sepsis (LOS), and death in infants born preterm. STUDY DESIGN Secondary data analysis was conducted via an extant cohort of 580 infants born <32 weeks of gestation and enrolled in 3 level III neonatal intensive care units. Prenatal antibiotic exposure was defined as antibiotics received by the mother within 72 hours before delivery. Postnatal empiric antibiotic exposure was defined as antibiotic initiated within the first day of life without documented infection, categorized as low (<5 days) or high (>5 days) duration. RESULTS Two-thirds of mothers received antibiotics within 72 hours before delivery, of whom 59.8% received >1 antibiotic. Ampicillin (37.6%) and azithromycin (26.4%) were the most common antibiotics given. NEC occurred in 7.5%, LOS in 11.1%, death in 9.6%, and the combined outcome of NEC, LOS, or death in 21.3% of study infants. In multiple logistic regression models adjusted for gestational age, postnatal empiric antibiotic exposure, and other factors, prenatal antibiotic exposure was associated with reduced risk of NEC (OR 0.28; 95% CI 0.14-0.56; P < .001), death (OR 0.29; 95% CI 0.14-0.60; P = .001), but not LOS (OR 1.59; 95% CI 0.84-2.99; P = .15), although protection was significant for the combined outcome (OR 0.52, P < .001). High postnatal empiric antibiotic exposure was associated with greater risk of death but not other outcomes in multiple regression models (OR 3.18, P = .002). CONCLUSIONS Prenatal antibiotic exposure was associated with lower rates of NEC or death of infants born preterm, and its impact on infant outcomes warrants further study.
Collapse
Affiliation(s)
- Benjamin D. Reed
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Kurt R. Schibler
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Hitesh Deshmukh
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Namasivayam Ambalavanan
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Ardythe L. Morrow
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA,Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA,Division of Biostatistics and Epidemiology, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
421
|
Makri V, Davies G, Cannell S, Willson K, Winterson L, Webb J, Kandhari A, Mansour M, Thomas J, Morris G, Matthes J, Banerjee S. Managing antibiotics wisely: a quality improvement programme in a tertiary neonatal unit in the UK. BMJ Open Qual 2018; 7:e000285. [PMID: 29756072 PMCID: PMC5942449 DOI: 10.1136/bmjoq-2017-000285] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 03/22/2018] [Accepted: 04/18/2018] [Indexed: 12/22/2022] Open
Abstract
Microbial resistance to antibiotics is a serious global health problem compounded by antibiotic overuse and limited investment in new antibiotic research. Inappropriate perinatal antibiotic exposure is increasingly linked to lifelong adverse outcomes through its impact on the developing microbiome. Antibiotic stewardship may be the only effective preventative strategy currently available. As the first tertiary neonatal unit in the UK to collaborate in an international quality improvement programme (QIP) with Vermont Oxford Network (VON), we present the results of our antibiotic stewardship initiative. The QIP was officially launched in January 2016 and aimed to reduce antibiotic usage rate (AUR) by 20% of baseline by 31st December 2016 without compromising patient safety. A multidisciplinary team of professionals and parent representatives shared good practices and improvement strategies through international webinars and local meetings, devised uniform data collection methodology and implemented a number of carefully selected ‘Plan–Do–Study–Act’ cycles. Run charts were used to present data and, where appropriate, statistical analysis undertaken to compare outcomes. The QIP resulted in a sustained reduction in AUR from a baseline median of 347 to 198 per 1000 patient-days (a reduction of 43%). The proportion of culture-negative sepsis screens where antibiotics were stopped within 36–48 hours increased consistently from a baseline of 32.5% to 91%. The antibiotic days per patient at discharge reduced from a median of 3 to 2 days, and there was a reduction in practice variation. Our annual mortality and necrotising enterocolitis rates for the VON cohort (<30 weeks or <1500 g) were the best ever recorded, 5.5% and 1.4%, respectively. Audits confirmed a high level of staff and family awareness of the QIP. The QIP achieved a sustained reduction in antibiotic use without compromising patient safety. Our challenge is to sustain this improvement safely.
Collapse
Affiliation(s)
- Vasiliki Makri
- Neonatal Unit, Abertawe Bro Morgannwg University Health Board, Swansea, UK
| | - Gemma Davies
- Neonatal Unit, Abertawe Bro Morgannwg University Health Board, Swansea, UK
| | - Stephanie Cannell
- Neonatal Unit, Abertawe Bro Morgannwg University Health Board, Swansea, UK
| | - Katherine Willson
- Neonatal Unit, Abertawe Bro Morgannwg University Health Board, Swansea, UK
| | - Lucy Winterson
- Neonatal Unit, Abertawe Bro Morgannwg University Health Board, Swansea, UK
| | - Joanna Webb
- Neonatal Unit, Abertawe Bro Morgannwg University Health Board, Swansea, UK
| | - Amit Kandhari
- Neonatal Unit, Abertawe Bro Morgannwg University Health Board, Swansea, UK
| | - Maha Mansour
- Neonatal Unit, Abertawe Bro Morgannwg University Health Board, Swansea, UK
| | - Joanne Thomas
- Neonatal Unit, Abertawe Bro Morgannwg University Health Board, Swansea, UK
| | - Geraint Morris
- Neonatal Unit, Abertawe Bro Morgannwg University Health Board, Swansea, UK
| | - Jean Matthes
- Neonatal Unit, Abertawe Bro Morgannwg University Health Board, Swansea, UK
| | - Sujoy Banerjee
- Neonatal Unit, Abertawe Bro Morgannwg University Health Board, Swansea, UK
| |
Collapse
|
422
|
Stinson LF, Payne MS, Keelan JA. A Critical Review of the Bacterial Baptism Hypothesis and the Impact of Cesarean Delivery on the Infant Microbiome. Front Med (Lausanne) 2018; 5:135. [PMID: 29780807 PMCID: PMC5945806 DOI: 10.3389/fmed.2018.00135] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 04/20/2018] [Indexed: 12/18/2022] Open
Abstract
Numerous studies suggest that infants delivered by cesarean section are at a greater risk of non-communicable diseases than their vaginal counterparts. In particular, epidemiological studies have linked Cesarean delivery with increased rates of asthma, allergies, autoimmune disorders, and obesity. Mode of delivery has also been associated with differences in the infant microbiome. It has been suggested that these differences are attributable to the "bacterial baptism" of vaginal birth, which is bypassed in cesarean deliveries, and that the abnormal establishment of the early-life microbiome is the mediator of later-life adverse outcomes observed in cesarean delivered infants. This has led to the increasingly popular practice of "vaginal seeding": the iatrogenic transfer of vaginal microbiota to the neonate to promote establishment of a "normal" infant microbiome. In this review, we summarize and critically appraise the current evidence for a causal association between Cesarean delivery and neonatal dysbiosis. We suggest that, while Cesarean delivery is certainly associated with alterations in the infant microbiome, the lack of exposure to vaginal microbiota is unlikely to be a major contributing factor. Instead, it is likely that indication for Cesarean delivery, intrapartum antibiotic administration, absence of labor, differences in breastfeeding behaviors, maternal obesity, and gestational age are major drivers of the Cesarean delivery microbial phenotype. We, therefore, call into question the rationale for "vaginal seeding" and support calls for the halting of this practice until robust evidence of need, efficacy, and safety is available.
Collapse
Affiliation(s)
- Lisa F Stinson
- Division of Obstetrics and Gynaecology, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Matthew S Payne
- Division of Obstetrics and Gynaecology, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Jeffrey A Keelan
- Division of Obstetrics and Gynaecology, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, WA, Australia
| |
Collapse
|
423
|
Berkhout DJC, Niemarkt HJ, de Boer NKH, Benninga MA, de Meij TGJ. The potential of gut microbiota and fecal volatile organic compounds analysis as early diagnostic biomarker for necrotizing enterocolitis and sepsis in preterm infants. Expert Rev Gastroenterol Hepatol 2018; 12:457-470. [PMID: 29488419 DOI: 10.1080/17474124.2018.1446826] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Although the exact pathophysiological mechanisms of both necrotizing enterocolitis (NEC) and late-onset sepsis (LOS) in preterm infants are yet to be elucidated, evidence is emerging that the gut microbiota plays a key role in their pathophysiology. Areas covered: In this review, initial microbial colonization and factors influencing microbiota composition are discussed. For both NEC and LOS, an overview of studies investigating preclinical alterations in gut microbiota composition and fecal volatile organic compounds (VOCs) is provided. Fecal VOCs are considered to reflect not only gut microbiota composition, but also their metabolic activity and concurrent interaction with the host. Expert review: Heterogeneity in study protocols and applied analytical techniques hampers reliable comparison between outcomes of different microbiota studies, limiting the ability to draw firm conclusions. This dilemma is illustrated by the finding that study results often cannot be reproduced, or even contradict each other. A NEC- and sepsis specific microbial or metabolic signature has not yet been discovered. Identification of 'disease-specific' VOCs and microbiota composition may increase understanding on pathophysiological mechanisms and may allow for development of an accurate screening tool, opening avenues towards timely identification and initiation of targeted treatment for preterm infants at increased risk for NEC and sepsis.
Collapse
Affiliation(s)
- Daniel Johannes Cornelis Berkhout
- a Department of Pediatric Gastroenterology , Emma Children's Hospital/Academic Medical Center , Amsterdam , the Netherlands.,b Department of Pediatric Gastroenterology , VU University Medical Center , Amsterdam , the Netherlands
| | | | - Nanne Klaas Hendrik de Boer
- d Department of Gastroenterology and Hepatology , VU University Medical Center , Amsterdam , the Netherlands
| | - Marc Alexander Benninga
- a Department of Pediatric Gastroenterology , Emma Children's Hospital/Academic Medical Center , Amsterdam , the Netherlands
| | | |
Collapse
|
424
|
Hospitalized Premature Infants Are Colonized by Related Bacterial Strains with Distinct Proteomic Profiles. mBio 2018; 9:mBio.00441-18. [PMID: 29636439 PMCID: PMC5893878 DOI: 10.1128/mbio.00441-18] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
During the first weeks of life, microbial colonization of the gut impacts human immune system maturation and other developmental processes. In premature infants, aberrant colonization has been implicated in the onset of necrotizing enterocolitis (NEC), a life-threatening intestinal disease. To study the premature infant gut colonization process, genome-resolved metagenomics was conducted on 343 fecal samples collected during the first 3 months of life from 35 premature infants housed in a neonatal intensive care unit, 14 of whom developed NEC, and metaproteomic measurements were made on 87 samples. Microbial community composition and proteomic profiles remained relatively stable on the time scale of a week, but the proteome was more variable. Although genetically similar organisms colonized many infants, most infants were colonized by distinct strains with metabolic profiles that could be distinguished using metaproteomics. Microbiome composition correlated with infant, antibiotics administration, and NEC diagnosis. Communities were found to cluster into seven primary types, and community type switched within infants, sometimes multiple times. Interestingly, some communities sampled from the same infant at subsequent time points clustered with those of other infants. In some cases, switches preceded onset of NEC; however, no species or community type could account for NEC across the majority of infants. In addition to a correlation of protein abundances with organism replication rates, we found that organism proteomes correlated with overall community composition. Thus, this genome-resolved proteomics study demonstrated that the contributions of individual organisms to microbiome development depend on microbial community context.IMPORTANCE Humans are colonized by microbes at birth, a process that is important to health and development. However, much remains to be known about the fine-scale microbial dynamics that occur during the colonization period. We conducted a genome-resolved study of microbial community composition, replication rates, and proteomes during the first 3 months of life of both healthy and sick premature infants. Infants were found to be colonized by similar microbes, but each underwent a distinct colonization trajectory. Interestingly, related microbes colonizing different infants were found to have distinct proteomes, indicating that microbiome function is not only driven by which organisms are present, but also largely depends on microbial responses to the unique set of physiological conditions in the infant gut.
Collapse
|
425
|
Stiemsma LT, Michels KB. The Role of the Microbiome in the Developmental Origins of Health and Disease. Pediatrics 2018; 141:e20172437. [PMID: 29519955 PMCID: PMC5869344 DOI: 10.1542/peds.2017-2437] [Citation(s) in RCA: 214] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/29/2017] [Indexed: 12/15/2022] Open
Abstract
Although the prominent role of the microbiome in human health has been established, the early-life microbiome is now being recognized as a major influence on long-term human health and development. Variations in the composition and functional potential of the early-life microbiome are the result of lifestyle factors, such as mode of birth, breastfeeding, diet, and antibiotic usage. In addition, variations in the composition of the early-life microbiome have been associated with specific disease outcomes, such as asthma, obesity, and neurodevelopmental disorders. This points toward this bacterial consortium as a mediator between early lifestyle factors and health and disease. In addition, variations in the microbial intrauterine environment may predispose neonates to specific health outcomes later in life. A role of the microbiome in the Developmental Origins of Health and Disease is supported in this collective research. Highlighting the early-life critical window of susceptibility associated with microbiome development, we discuss infant microbial colonization, beginning with the maternal-to-fetal exchange of microbes in utero and up through the influence of breastfeeding in the first year of life. In addition, we review the available disease-specific evidence pointing toward the microbiome as a mechanistic mediator in the Developmental Origins of Health and Disease.
Collapse
Affiliation(s)
- Leah T Stiemsma
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, California
| | - Karin B Michels
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, California
| |
Collapse
|
426
|
Clostridial Strain-Specific Characteristics Associated with Necrotizing Enterocolitis. Appl Environ Microbiol 2018; 84:AEM.02428-17. [PMID: 29352082 DOI: 10.1128/aem.02428-17] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 01/11/2018] [Indexed: 12/21/2022] Open
Abstract
We aimed at identifying potential bacterial factors linking clostridia with necrotizing enterocolitis (NEC). We compared the phenotypic traits, stress responses, cellular cytotoxicity, and inflammatory capabilities of the largest collection of Clostridium butyricum and Clostridium neonatale strains isolated from fecal samples of NEC preterm neonates (PN) and control PNs. When strain characteristics were used as explanatory variables, a statistical discriminant analysis allowed the separation of NEC and control strains into separate groups. Strains isolated from NEC PN were characterized by a higher viability at 30°C (P = 0.03) and higher aerotolerance (P = 0.01), suggesting that NEC strains may have a competitive and/or survival advantage in the environmental gastrointestinal tract conditions of NEC PN. Heat-treated NEC bacteria induced higher production of interleukin-8 in Caco-2 cells (P = 0.03), suggesting proinflammatory activity. In vitro, bacteria, bacterial components, and fecal filtrates showed variable cytotoxic effects affecting the cellular network and/or cell viability, without specific association with NEC or control samples. Altogether, our data support the existence of a specific clostridial strain signature associated with NEC.IMPORTANCE Clostridia are part of the commensal microbiota in preterm neonates (PN). However, microbiota analyses by culture and metagenomics have linked necrotizing enterocolitis (NEC) and intestinal colonization with clostridial species. Nevertheless, little is known about the specific characteristics that may be shared by clostridia associated with NEC compared to commensal clostridia. Therefore, our goal was to identify specific bacterial factors linking clostridial strains with NEC. We report the existence of a specific bacterial signature associated with NEC and propose that activation of the innate immune response may be a unifying causative mechanism for the development of NEC independent of a specific pathogenic organism. The present study provides new insights into NEC pathophysiology that are needed for better diagnostics and strategies for implementing prevention of the disease.
Collapse
|
427
|
Denning NL, Prince JM. Neonatal intestinal dysbiosis in necrotizing enterocolitis. Mol Med 2018; 24:4. [PMID: 30134786 PMCID: PMC6016883 DOI: 10.1186/s10020-018-0002-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 02/13/2018] [Indexed: 12/11/2022] Open
Abstract
Necrotizing Enterocolitis (NEC) is one of the most devastating gastrointestinal diseases in neonates, particularly among preterm infants in whom surgical NEC is the leading cause of morbidity. NEC pathophysiology occurs in the hyper-reactive milieu of the premature gut after bacterial colonization. The resultant activation of the TLR4 pathway appears to be a strongly contributing factor. Advancements in metagenomics may yield new clarity to the relationship between the neonatal intestinal microbiome and the development of NEC. After a century without effective directed treatments, microbiome manipulation offers a promising therapeutic target for the prevention and treatment of this devastating disease.
Collapse
Affiliation(s)
- Naomi-Liza Denning
- Division of Pediatric Surgery, Zucker School of Medicine at Hofstra/Northwell, Cohen Children's Medical Center, 269-01 76th Avenue, CH 158, New Hyde Park, New York, NY, 11040, USA. .,Feinstein Institute for Medical Research, Manhasset, NY, 11030, USA.
| | - Jose M Prince
- Division of Pediatric Surgery, Zucker School of Medicine at Hofstra/Northwell, Cohen Children's Medical Center, 269-01 76th Avenue, CH 158, New Hyde Park, New York, NY, 11040, USA.,Feinstein Institute for Medical Research, Manhasset, NY, 11030, USA.,Trauma Institute, Northwell Health System, Manhasset, NY, 11030, USA
| |
Collapse
|
428
|
Splichalova A, Slavikova V, Splichalova Z, Splichal I. Preterm Life in Sterile Conditions: A Study on Preterm, Germ-Free Piglets. Front Immunol 2018; 9:220. [PMID: 29491864 PMCID: PMC5817058 DOI: 10.3389/fimmu.2018.00220] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 01/25/2018] [Indexed: 12/21/2022] Open
Abstract
Preterm infants born with immature organ systems, which can impede normal development, can also be highly sensitive to different biological and/or environmental factors. Animal models could aid in investigating and understanding the effects of different conditions on the health of these immunocompromised infants. The epitheliochorial placentation of the pig prevents the prenatal transfer of protective colostral immunoglobulins. Surgical colostrum-deprived piglets are free of maternal immunoglobulins, and the cells that are normally provided via colostrum. We bred preterm germ-free piglets in sterile conditions and compared them with their term counterparts. Enterocyte development and intestinal morphology, tight junction proteins claudin-1 and occludin, pattern-recognizing receptors, adaptor molecules and coreceptors (RAGE, TLR2, TLR4, TLR9, MyD88, TRIF, MD2, and CD14), and inflammasome NLRP3 transcription were all evaluated. The production of inflammatory mediators IFN-α, IL-4, IL-6, IL-8, IL-10, IL-12/23 p40, TNF-α, IFN-γ, and high mobility group box 1 (HMGB1) in the intestine of germ-free piglets was also assessed. In the preterm germ-free piglets, the ileum showed decreased lamina propria cellularity, reduced villous height, and thinner and less distinct stratification - especially muscle layer, in comparison with their term counterparts. Claudin-1 transcription increased in the intestine of the preterm piglets. The transcription levels of pattern-recognizing receptors and adaptor molecules showed ambiguous trends between the groups. The levels of IL-6, IL-8, IL-10, and TNF-α were increased in the preterm ileum numerically (though not significantly), with statistically significant increases in the colon. Additionally, IL-12/23 p40 and IFN-γ were statistically significantly higher in the preterm colon. Both blood plasma and intestinal HMGB1 levels were nonsignificantly higher in the preterm group. We propose that the intestine of the preterm germ-free piglets showed "mild inflammation in sterile conditions." This model, which establishes preterm, hysterectomy-derived germ-free piglets, without protective maternal immunoglobulins, can be used to study influences of microbiota, nutrition, and therapeutic interventions on the development and health of vulnerable immunocompromised preterm infants.
Collapse
Affiliation(s)
- Alla Splichalova
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Novy Hradek, Czechia
| | - Vera Slavikova
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Novy Hradek, Czechia
| | - Zdislava Splichalova
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Novy Hradek, Czechia
| | - Igor Splichal
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Novy Hradek, Czechia
| |
Collapse
|
429
|
Abstract
In this review, we summarize existing knowledge regarding the effects of probiotics on necrotizing enterocolitis (NEC). We review the role of the microbiome in NEC and pre-clinical data on mechanisms of probiotic action. Next, we summarize existing randomized controlled trials and observational studies of probiotics to prevent NEC. We also summarize findings from several recent meta-analyses and report a new cumulative meta-analysis of probiotic trials. Finally, we review data from cohorts routinely using commercially available probiotics. Our goal is to inform clinicians about the risks and benefits of probiotics, which may be helpful for those considering use in preterm infants to prevent NEC, death, or sepsis.
Collapse
Affiliation(s)
- Ravi Mangal Patel
- Department of Pediatrics, Division of Neonatology, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Dr. NE, 3rd Floor, Atlanta, Georgia 30322.
| | - Mark A. Underwood
- Professor, Department of Pediatrics, Division of Neonatology, University of California Davis School of Medicine, Sacramento, CA
| |
Collapse
|
430
|
Abstract
PURPOSE OF REVIEW Emerging research on the pediatric microbiome implicates the importance of the microbiome on the development of the immune system, nervous system, and growth. Changes to the microbiome during infancy are associated with the development of chronic illnesses such as asthma and inflammatory bowel disease. Additionally, the microbiome provides protection against certain pathogens, affects vaccine responses, and alters drug metabolism. This review highlights what is known about the microbiome, the establishment of a healthy microbiome and the significance that changes to the microbiome composition have on growth and health of children and adolescents. RECENT FINDINGS Vaginal delivery, breastfeeding, maternal health, and nutrition help shape a healthy microbiome. Caesarian delivery, formula feeding, and antibiotic use perturb the microbiome and are associated with the development of type II diabetes, asthma, allergic diseases, and obesity later in life. Specific interventions using pre and probiotics in multiple settings are under investigation with limited success. SUMMARY A better understanding of the microbiome and the interaction with the immune system may help guide interventions to alter the microbiome toward a state of lifelong health.
Collapse
|
431
|
Abstract
Necrotizing enterocolitis (NEC) continues to afflict approximately 7% of preterm infants born weighing less than 1500g, though recent investigations have provided novel insights into the pathogenesis of this complex disease. The disease has been a major cause of morbidity and mortality in neonatal intensive care units worldwide for many years, and our current understanding reflects exceptional observations made decades ago. In this review, we will describe NEC from a historical context and summarize seminal findings that underscore the importance of enteral feeding, the gut microbiota, and intestinal inflammation in this complex pathophysiology.
Collapse
Affiliation(s)
- David Hackam
- Division of Pediatric General Surgery, Department of Surgery, Johns Hopkins Children's Center and The Johns Hopkins University, Baltimore, MD.
| | - Michael Caplan
- North Shore University Health System and the University of Chicago Pritzker School of Medicine
| |
Collapse
|
432
|
Le Bastard Q, Al-Ghalith GA, Grégoire M, Chapelet G, Javaudin F, Dailly E, Batard E, Knights D, Montassier E. Systematic review: human gut dysbiosis induced by non-antibiotic prescription medications. Aliment Pharmacol Ther 2018; 47:332-345. [PMID: 29205415 DOI: 10.1111/apt.14451] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 09/04/2017] [Accepted: 11/14/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Global prescription drug use has been increasing continuously for decades. The gut microbiome, a key contributor to health status, can be altered by prescription drug use, as antibiotics have been repeatedly described to have both short-term and long-standing effects on the intestinal microbiome. AIM To summarise current findings on non-antibiotic prescription-induced gut microbiome changes, focusing on the most frequently prescribed therapeutic drug categories. METHODS We conducted a systematic review by first searching in online databases for indexed articles and abstracts in accordance with PRISMA guidelines. Studies assessing the intestinal microbiome alterations associated with proton pump inhibitors (PPIs), metformin, nonsteroidal anti-inflammatory drugs (NSAIDs), opioids, statins and antipsychotics were included. We only included studies using culture-independent molecular techniques. RESULTS Proton pump inhibitors and antipsychotic medications are associated with a decrease in α diversity in the gut microbiome, whereas opioids were associated with an increase in α diversity. Metformin and NSAIDs were not associated with significant changes in α diversity. β diversity was found to be significantly altered with all drugs, except for NSAIDs. PPI use was linked to a decrease in Clotridiales and increase in Actinomycetales, Micrococcaceae and Streptococcaceae, which are changes previously implicated in dysbiosis and increased susceptibility to Clostridium difficile infection. Consistent results showed that PPIs, metformin, NSAIDs, opioids and antipsychotics were either associated with increases in members of class Gammaproteobacteria (including Enterobacter, Escherichia, Klebsiella and Citrobacter), or members of family Enterococcaceae, which are often pathogens isolated from bloodstream infections in critically ill patients. We also found that antipsychotic treatment, usually associated with an increase in body mass index, was marked by a decreased ratio of Bacteroidetes:Firmicutes in the gut microbiome, resembling trends seen in obese patients. CONCLUSIONS Non-antibiotic prescription drugs have a notable impact on the overall architecture of the intestinal microbiome. Further explorations should seek to define biomarkers of dysbiosis induced by specific drugs, and potentially tailor live biotherapeutics to counter this drug-induced dysbiosis. Many other frequently prescribed drugs should also be investigated to better understand the link between these drugs, the microbiome and health status.
Collapse
Affiliation(s)
- Q Le Bastard
- MiHAR Lab, Institut de Recherche en Santé 2, Université de Nantes, Nantes, France
| | - G A Al-Ghalith
- Biotechnology Institute, University of Minnesota, Saint Paul, MN, USA.,Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, USA
| | - M Grégoire
- MiHAR Lab, Institut de Recherche en Santé 2, Université de Nantes, Nantes, France
| | - G Chapelet
- MiHAR Lab, Institut de Recherche en Santé 2, Université de Nantes, Nantes, France
| | - F Javaudin
- MiHAR Lab, Institut de Recherche en Santé 2, Université de Nantes, Nantes, France
| | - E Dailly
- MiHAR Lab, Institut de Recherche en Santé 2, Université de Nantes, Nantes, France
| | - E Batard
- MiHAR Lab, Institut de Recherche en Santé 2, Université de Nantes, Nantes, France
| | - D Knights
- Biotechnology Institute, University of Minnesota, Saint Paul, MN, USA.,Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, USA
| | - E Montassier
- MiHAR Lab, Institut de Recherche en Santé 2, Université de Nantes, Nantes, France
| |
Collapse
|
433
|
Itani T, Ayoub Moubareck C, Melki I, Rousseau C, Mangin I, Butel MJ, Karam-Sarkis D. Preterm infants with necrotising enterocolitis demonstrate an unbalanced gut microbiota. Acta Paediatr 2018; 107:40-47. [PMID: 28921627 DOI: 10.1111/apa.14078] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 07/21/2017] [Accepted: 09/13/2017] [Indexed: 02/04/2023]
Abstract
AIM This Lebanese study tested the hypothesis that differences would exist in the gut microbiota of preterm infants with and without necrotising enterocolitis (NEC), as reported in Western countries. METHODS This study compared 11 infants with NEC and 11 controls, all born at 27-35 weeks, in three neonatal intensive care units between January 2013 and March 2015. Faecal samples were collected at key time points, and microbiota was analysed by culture, quantitative PCR (qPCR) and temperature temporal gel electrophoresis (TTGE). RESULTS The cultures revealed that all preterm infants were poorly colonised and harboured no more than seven species. Prior to NEC diagnosis, significant differences were observed by qPCR with a higher colonisation by staphylococci (p = 0.034) and lower colonisations by enterococci (p = 0.039) and lactobacilli (p = 0.048) in the NEC group compared to the healthy controls. Throughout the study, virtually all of the infants were colonised by Enterobacteriaceae at high levels. TTGE analysis revealed no particular clusterisation, showing high interindividual variability. CONCLUSION The NEC infants were poorly colonised with no more than seven species, and the controls had a more diversified and balanced gut microbiota. Understanding NEC aetiology better could lead to more effective prophylactic interventions and a reduced incidence.
Collapse
Affiliation(s)
- Tarek Itani
- Microbiology Laboratory; Faculty of Pharmacy; Saint-Joseph University; Beirut Lebanon
| | - Carole Ayoub Moubareck
- Microbiology Laboratory; Faculty of Pharmacy; Saint-Joseph University; Beirut Lebanon
- College of Natural and Health Sciences; Zayed University; Dubai UAE
| | - Imad Melki
- Hôtel Dieu de France Hospital; Beirut Lebanon
| | - Clotilde Rousseau
- Department Risk in pregnancy; Hospital University; Paris Descartes University; Sorbonne Paris Cité; Paris France
- Microbiology; Saint-Louis Hospital; APHP; Paris France
| | - Irène Mangin
- Department Risk in pregnancy; Hospital University; Paris Descartes University; Sorbonne Paris Cité; Paris France
- Laboratoire MIEL; Conservatoire national des arts et métiers; Paris France
| | - Marie-José Butel
- Department Risk in pregnancy; Hospital University; Paris Descartes University; Sorbonne Paris Cité; Paris France
| | - Dolla Karam-Sarkis
- Microbiology Laboratory; Faculty of Pharmacy; Saint-Joseph University; Beirut Lebanon
| |
Collapse
|
434
|
Esaiassen E, Hjerde E, Cavanagh JP, Pedersen T, Andresen JH, Rettedal SI, Støen R, Nakstad B, Willassen NP, Klingenberg C. Effects of Probiotic Supplementation on the Gut Microbiota and Antibiotic Resistome Development in Preterm Infants. Front Pediatr 2018; 6:347. [PMID: 30505830 PMCID: PMC6250747 DOI: 10.3389/fped.2018.00347] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/26/2018] [Indexed: 12/15/2022] Open
Abstract
Objectives: In 2014 probiotic supplementation (Lactobacillus acidophilus and Bifidobacterium longum subspecies infantis; InfloranⓇ) was introduced as standard of care to prevent necrotizing enterocolitis (NEC) in extremely preterm infants in Norway. We aimed to evaluate the influence of probiotics and antibiotic therapy on the developing gut microbiota and antibiotic resistome in extremely preterm infants, and to compare with very preterm infants and term infants not given probiotics. Study design: A prospective, observational multicenter study in six tertiary-care neonatal units. We enrolled 76 infants; 31 probiotic-supplemented extremely preterm infants <28 weeks gestation, 35 very preterm infants 28-31 weeks gestation not given probiotics and 10 healthy full-term control infants. Taxonomic composition and collection of antibiotic resistance genes (resistome) in fecal samples, collected at 7 and 28 days and 4 months age, were analyzed using shotgun-metagenome sequencing. Results: Median (IQR) birth weight was 835 (680-945) g and 1,290 (1,150-1,445) g in preterm infants exposed and not exposed to probiotics, respectively. Two extremely preterm infants receiving probiotic developed NEC requiring surgery. At 7 days of age we found higher median relative abundance of Bifidobacterium in probiotic supplemented infants (64.7%) compared to non-supplemented preterm infants (0.0%) and term control infants (43.9%). Lactobacillus was only detected in small amounts in all groups, but the relative abundance increased up to 4 months. Extremely preterm infants receiving probiotics had also much higher antibiotic exposure, still overall microbial diversity and resistome was not different than in more mature infants at 4 weeks and 4 months. Conclusion: Probiotic supplementation may induce colonization resistance and alleviate harmful effects of antibiotics on the gut microbiota and antibiotic resistome. Clinical Trial Registration: Clinicaltrials.gov: NCT02197468. https://clinicaltrials.gov/ct2/show/NCT02197468.
Collapse
Affiliation(s)
- Eirin Esaiassen
- Paediatric Research Group, Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway.,Department of Paediatrics, University Hospital of North Norway, Tromsø, Norway
| | - Erik Hjerde
- Department of Chemistry, Norstruct, UiT The Arctic University of Norway, Tromsø, Norway
| | - Jorunn Pauline Cavanagh
- Paediatric Research Group, Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway.,Department of Paediatrics, University Hospital of North Norway, Tromsø, Norway
| | - Tanja Pedersen
- Department of Paediatrics, Haukeland University Hospital, Bergen, Norway
| | - Jannicke H Andresen
- Department of Neonatal Intensive Care, Oslo University Hospital, Oslo, Norway
| | - Siren I Rettedal
- Department of Paediatrics, Stavanger University Hospital, Stavanger, Norway
| | - Ragnhild Støen
- Department of Paediatrics, St. Olavs University Hospital, Trondheim, Norway.,Department of Laboratory Medicine, Children's and Women's Health, University of Science and Technology, Trondheim, Norway
| | - Britt Nakstad
- Department of Paediatric and Adolescents Medicine, Akershus University Hospital, Nordbyhagen, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Nils P Willassen
- Department of Chemistry, Norstruct, UiT The Arctic University of Norway, Tromsø, Norway
| | - Claus Klingenberg
- Paediatric Research Group, Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway.,Department of Paediatrics, University Hospital of North Norway, Tromsø, Norway
| |
Collapse
|
435
|
Dobbler PT, Procianoy RS, Mai V, Silveira RC, Corso AL, Rojas BS, Roesch LFW. Low Microbial Diversity and Abnormal Microbial Succession Is Associated with Necrotizing Enterocolitis in Preterm Infants. Front Microbiol 2017; 8:2243. [PMID: 29187842 PMCID: PMC5695202 DOI: 10.3389/fmicb.2017.02243] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 10/31/2017] [Indexed: 12/15/2022] Open
Abstract
Despite increased efforts, the diverse etiologies of Necrotizing Enterocolitis (NEC) have remained largely elusive. Clinical predictors of NEC remain ill-defined and currently lack sufficient specificity. The development of a thorough understanding of initial gut microbiota colonization pattern in preterm infants might help to improve early detection or prediction of NEC and its associated morbidities. Here we compared the fecal microbiota successions, microbial diversity, abundance and structure of newborns that developed NEC with preterm controls. A 16S rRNA based microbiota analysis was conducted in a total of 132 fecal samples that included the first stool (meconium) up until the 5th week of life or NEC diagnosis from 40 preterm babies (29 controls and 11 NEC cases). A single phylotype matching closest to the Enterobacteriaceae family correlated strongly with NEC. In DNA from the sample with the greatest abundance of this phylotype additional shotgun metagenomic sequencing revealed Citrobacter koseri and Klebsiella pneumoniae as the dominating taxa. These two taxa might represent suitable microbial biomarker targets for early diagnosis of NEC. In NEC cases, we further detected lower microbial diversity and an abnormal succession of the microbial community before NEC diagnosis. Finally, we also detected a disruption in anaerobic microorganisms in the co-occurrence network of meconium samples from NEC cases. Our data suggest that a strong dominance of Citrobacter koseri and/or Klebsiella pneumoniae, low diversity, low abundance of Lactobacillus, as well as an altered microbial-network structure during the first days of life, correlate with NEC risk in preterm infants. Confirmation of these findings in other hospitals might facilitate the development of a microbiota based screening approach for early detection of NEC.
Collapse
Affiliation(s)
- Priscila T Dobbler
- Centro Interdisciplinar de Pesquisas em Biotecnologia - CIP-Biotec, Campus São Gabriel, Universidade Federal do Pampa, São Gabriel, Brazil
| | - Renato S Procianoy
- Serviço de Neonatologia do Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Volker Mai
- Department of Epidemiology, College of Public Health and Health Professions and College of Medicine, Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
| | - Rita C Silveira
- Serviço de Neonatologia do Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Andréa L Corso
- Serviço de Neonatologia do Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Bruna S Rojas
- Serviço de Neonatologia do Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Luiz F W Roesch
- Centro Interdisciplinar de Pesquisas em Biotecnologia - CIP-Biotec, Campus São Gabriel, Universidade Federal do Pampa, São Gabriel, Brazil
| |
Collapse
|
436
|
Kolter J, Henneke P. Codevelopment of Microbiota and Innate Immunity and the Risk for Group B Streptococcal Disease. Front Immunol 2017; 8:1497. [PMID: 29209311 PMCID: PMC5701622 DOI: 10.3389/fimmu.2017.01497] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 10/24/2017] [Indexed: 12/14/2022] Open
Abstract
The pathogenesis of neonatal late-onset sepsis (LOD), which manifests between the third day and the third month of life, remains poorly understood. Group B Streptococcus (GBS) is the most important cause of LOD in infants without underlying diseases or prematurity and the third most frequent cause of meningitis in the Western world. On the other hand, GBS is a common intestinal colonizer in infants. Accordingly, despite its adaption to the human lower gastrointestinal tract, GBS has retained its potential virulence and its transition from a commensal to a dangerous pathogen is unpredictable in the individual. Several cellular innate immune mechanisms, in particular Toll-like receptors, the inflammasome and the cGAS pathway, are engaged by GBS effectors like nucleic acids. These are likely to impact on the GBS-specific host resistance. Given the long evolution of streptococci as a normal constituent of the human microbiota, the emergence of GBS as the dominant neonatal sepsis cause just about 50 years ago is remarkable. It appears that intensive usage of tetracycline starting in the 1940s has been a selection advantage for the currently dominant GBS clones with superior adhesive and invasive properties. The historical replacement of Group A by Group B streptococci as a leading neonatal pathogen and the higher frequency of other β-hemolytic streptococci in areas with low GBS prevalence suggests the existence of a confined streptococcal niche, where locally competing streptococcal species are subject to environmental and immunological selection pressure. Thus, it seems pivotal to resolve neonatal innate immunity at mucous surfaces and its impact on microbiome composition and quality, i.e., genetic heterogeneity and metabolism, at the microanatomical level. Then, designer pro- and prebiotics, such as attenuated strains of GBS, and oligonucleotide priming of mucosal immunity may unfold their potential and facilitate adaptation of potentially hazardous streptococci as part of a beneficial local microbiome, which is stabilized by mucocutaneous innate immunity.
Collapse
Affiliation(s)
- Julia Kolter
- Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Philipp Henneke
- Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Pediatrics and Adolescent Medicine, Medical Center - University of Freiburg, Freiburg, Germany
| |
Collapse
|
437
|
Current Resources for Evidence-Based Practice, November/December 2017. J Obstet Gynecol Neonatal Nurs 2017; 46:878-885. [DOI: 10.1016/j.jogn.2017.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
438
|
Choi YS, Song IG. Fetal and preterm infant microbiomes: a new perspective of necrotizing enterocolitis. KOREAN JOURNAL OF PEDIATRICS 2017; 60:307-311. [PMID: 29158764 PMCID: PMC5687977 DOI: 10.3345/kjp.2017.60.10.307] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 08/13/2017] [Accepted: 09/17/2017] [Indexed: 12/19/2022]
Abstract
Necrotizing enterocolitis (NEC) is a devastating condition of hospitalized preterm infants. Numerous studies have attempted to identify the cause of NEC by examining the immunological features associated with pathogenic microorganisms. No single organism has proven responsible for the disease; however, immunological studies are now focused on the microbiome. Recent research has investigated the numerous bacterial species residing in the body and their role in diseases in preterm infants. The timing of initial microbial colonization is a subject of interest. The microbiome appears to transfer from the mother to the newborn, as well as to the fetus. Cross-talk between the fetus and fetal microbiome takes place continuously to generate a unique immune system. This review examined the transfer of the microbiome to the human fetus, and its potential relationship with NEC.
Collapse
Affiliation(s)
- Yong-Sung Choi
- Department of Pediatrics, Kyung Hee University School of Medicine, Seoul, Korea
| | - In Gyu Song
- Department of Pediatrics, Kyung Hee University School of Medicine, Seoul, Korea
| |
Collapse
|
439
|
Sohn K, Underwood MA. Prenatal and postnatal administration of prebiotics and probiotics. Semin Fetal Neonatal Med 2017; 22:284-289. [PMID: 28720399 PMCID: PMC5618799 DOI: 10.1016/j.siny.2017.07.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Colonization of the neonatal gut by beneficial bacteria is important for the establishment and maintenance of the mucosal barrier, thus protecting the neonate from enteric pathogens and local and systemic inflammation. The neonatal microbiome is influenced by infant diet, environment, and the maternal microbiome. Dysbiosis in pregnancy increases the risk of pre-eclampsia, diabetes, infection, preterm labor, and later childhood atopy. Dysbiosis of the neonatal gut plays an important role in colic in the term infant, in the disease processes which plague preterm infants, including necrotizing enterocolitis and sepsis, and in the long-term outcomes of neonates. Administration of enteral prebiotics, probiotics, and synbiotics during pregnancy, lactation, and postnatal life appears to be a safe and feasible method to alter the maternal and neonatal microbiome, thus improving pregnancy and neonatal outcomes.
Collapse
Affiliation(s)
| | - Mark A. Underwood
- Corresponding author. Address: Department of Pediatrics, University of California Davis School of Medicine, 2516 Stockton Blvd, Suite 253, Sacramento, CA 95817, USA. Tel.: +1 916 734 8672; fax: +1 916 456 4490. (M.A. Underwood)
| |
Collapse
|
440
|
Rusconi B, Good M, Warner BB. The Microbiome and Biomarkers for Necrotizing Enterocolitis: Are We Any Closer to Prediction? J Pediatr 2017; 189:40-47.e2. [PMID: 28669607 PMCID: PMC5614810 DOI: 10.1016/j.jpeds.2017.05.075] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 04/24/2017] [Accepted: 05/26/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Brigida Rusconi
- Division of Gastroenterology, Hepatology & Nutrition, Pathobiology Research Unit, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Misty Good
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Barbara B. Warner
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
441
|
Tam PKH, Chung PHY, St Peter SD, Gayer CP, Ford HR, Tam GCH, Wong KKY, Pakarinen MP, Davenport M. Advances in paediatric gastroenterology. Lancet 2017; 390:1072-1082. [PMID: 28901937 DOI: 10.1016/s0140-6736(17)32284-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/20/2017] [Accepted: 08/01/2017] [Indexed: 12/17/2022]
Abstract
Recent developments in paediatric gastrointestinal surgery have focused on minimally invasive surgery, the accumulation of high-quality clinical evidence, and scientific research. The benefits of minimally invasive surgery for common disorders like appendicitis and hypertrophic pyloric stenosis are all supported by good clinical evidence. Although minimally invasive surgery has been extended to neonatal surgery, it is difficult to establish its role for neonatal disorders such as oesophageal atresia and biliary atresia through clinical trials because of the rarity of these disorders. Advances in treatments for biliary atresia and necrotising enterocolitis have been achieved through specialisation, multidisciplinary management, and multicentre collaboration in research; similarly robust clinical evidence for other rare gastrointestinal disorders is needed. As more neonates with gastrointestinal diseases survive into adulthood, their long-term sequelae will also need evidence-based multidisciplinary care. Identifying cures for long-term problems of a complex developmental anomaly such as Hirschsprung's disease will rely on unravelling its pathogenesis through genetics and the development of stem-cell therapy.
Collapse
Affiliation(s)
- Paul K H Tam
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong.
| | - Patrick H Y Chung
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Shawn D St Peter
- Department of General & Thoracic Surgery, Children's Mercy Hospital and Clinics, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Christopher P Gayer
- Department of Surgery, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Henri R Ford
- Department of Surgery, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Greta C H Tam
- School of Public Health, Chinese University of Hong Kong, Hong Kong
| | - Kenneth K Y Wong
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Mikko P Pakarinen
- Paediatric Surgery and Paediatric Liver and Gut Research Group, Children's Hospital, University of Helsinki and Helsinki University Hospital, Finland
| | - Mark Davenport
- Department of Paediatric Surgery, King's College Hospital, London, UK
| |
Collapse
|
442
|
Rozé JC, Ancel PY, Lepage P, Martin-Marchand L, Al Nabhani Z, Delannoy J, Picaud JC, Lapillonne A, Aires J, Durox M, Darmaun D, Neu J, Butel MJ. Nutritional strategies and gut microbiota composition as risk factors for necrotizing enterocolitis in very-preterm infants. Am J Clin Nutr 2017; 106:821-830. [PMID: 28659297 PMCID: PMC5573022 DOI: 10.3945/ajcn.117.152967] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 06/05/2017] [Indexed: 12/22/2022] Open
Abstract
Background: The pathophysiology of necrotizing enterocolitis (NEC) remains poorly understood.Objective: We assessed the relation between feeding strategies, intestinal microbiota composition, and the development of NEC.Design: We performed a prospective nationwide population-based study, EPIPAGE 2 (Etude Epidémiologique sur les Petits Ages Gestationnels), including preterm infants born at <32 wk of gestation in France in 2011. From individual characteristics observed during the first week of life, we calculated a propensity score for the risk of NEC (Bell's stage 2 or 3) after day 7 of life. We analyzed the relation between neonatal intensive care unit (NICU) strategies concerning the rate of progression of enteral feeding, the direct-breastfeeding policy, and the onset of NEC using general linear mixed models to account for clustering by the NICU. An ancillary propensity-matched case-control study, EPIFLORE (Etude Epidémiologique de la flore), in 20 of the 64 NICUs, analyzed the intestinal microbiota by culture and 16S ribosomal RNA gene sequencing.Results: Among the 3161 enrolled preterm infants, 106 (3.4%; 95% CI: 2.8%, 4.0%) developed NEC. Individual characteristics were significantly associated with NEC. Slower and intermediate rates of progression of enteral feeding strategies were associated with a higher risk of NEC, with an adjusted OR of 2.3 (95% CI: 1.2, 4.5; P = 0.01) and 2.0 (95% CI: 1.1, 3.5; P = 0.02), respectively. Less favorable and intermediate direct-breastfeeding policies were associated with higher NEC risk as well, with an adjusted OR of 2.5 (95% CI: 1.1, 5.8; P = 0.03) and 2.3 (95% CI: 1.1, 4.8; P = 0.02), respectively. Microbiota analysis performed in 16 cases and 78 controls showed an association between Clostridium neonatale and Staphylococcus aureus with NEC (P = 0.001 and P = 0.002).Conclusions: A slow rate of progression of enteral feeding and a less favorable direct-breastfeeding policy are associated with an increased risk of developing NEC. For a given level of risk assessed by propensity score, colonization by C. neonatale and/or S. aureus is significantly associated with NEC. This trial (EPIFLORE study) was registered at clinicaltrials.gov as NCT01127698.
Collapse
Affiliation(s)
- Jean-Christophe Rozé
- Department of Neonatal Medicine, .,Epidémiologie Clinique, Clinical Investigation Center - Clinical Epidemiology (CIC004), and.,INRA, UMR 1280 Physiology of Nutritional Adaptations, Nantes University Hospital, Nantes, France
| | - Pierre-Yves Ancel
- INSERM, U1153, Obstetrical, Perinatal and Pediatric Epidemiology Team, Epidemiology and Statistics Sorbonne Paris Cité Research Center, Paris Descartes University, Paris, France,Risks in Pregnancy Department, Paris Descartes University, Paris, France,Clinical investigation center CIC P1419, Cochin Hotel-Dieu Hospital, AP-HP, Paris, France
| | - Patricia Lepage
- Micalis Institute, INRA, AgroParisTech, University Paris-Saclay, Paris, France
| | - Laetitia Martin-Marchand
- INSERM, U1153, Obstetrical, Perinatal and Pediatric Epidemiology Team, Epidemiology and Statistics Sorbonne Paris Cité Research Center, Paris Descartes University, Paris, France
| | - Ziad Al Nabhani
- Micalis Institute, INRA, AgroParisTech, University Paris-Saclay, Paris, France
| | - Johanne Delannoy
- Risks in Pregnancy Department, Paris Descartes University, Paris, France,EA 4065 Intestinal Ecosystem, Probiotics, Antibiotics, Faculty of Pharmacy, Paris Descartes University, Paris, France
| | - Jean-Charles Picaud
- Department of Neonatal Medicine, Croix Rousse Hospital, Lyon Hospitals, Lyon, France
| | - Alexandre Lapillonne
- Department of Neonatal Medicine, AP-HP, Necker Enfants Malades Hospital, Paris, France
| | - Julio Aires
- Risks in Pregnancy Department, Paris Descartes University, Paris, France,EA 4065 Intestinal Ecosystem, Probiotics, Antibiotics, Faculty of Pharmacy, Paris Descartes University, Paris, France
| | - Mélanie Durox
- INSERM, U1153, Obstetrical, Perinatal and Pediatric Epidemiology Team, Epidemiology and Statistics Sorbonne Paris Cité Research Center, Paris Descartes University, Paris, France
| | - Dominique Darmaun
- INRA, UMR 1280 Physiology of Nutritional Adaptations, Nantes University Hospital, Nantes, France
| | - Josef Neu
- Department of Pediatrics, University of Florida, Gainesville, FL
| | - Marie-José Butel
- Risks in Pregnancy Department, Paris Descartes University, Paris, France,EA 4065 Intestinal Ecosystem, Probiotics, Antibiotics, Faculty of Pharmacy, Paris Descartes University, Paris, France
| | | | | |
Collapse
|
443
|
Park AM, Omura S, Fujita M, Sato F, Tsunoda I. Helicobacter pylori and gut microbiota in multiple sclerosis versus Alzheimer's disease: 10 pitfalls of microbiome studies. CLINICAL & EXPERIMENTAL NEUROIMMUNOLOGY 2017; 8:215-232. [PMID: 29158778 DOI: 10.1111/cen3.12401] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Alteration of microbiota has been associated with intestinal, inflammatory, and neurological diseases. Abundance of "good bacteria" such as Bifidobacterium, or their products have been generally believed to be beneficial for any diseases, while "bad bacteria" such as pathogenic Helicobacter pylori are assumed to be always detrimental for hosts. However, this is not the case when we compare and contrast the association of the gut microbiota with two neurological diseases, multiple sclerosis (MS) and Alzheimer's disease (AD). Following H. pylori infection, pro-inflammatory T helper (Th)1 and Th17 immune response are initially induced to eradicate bacteria. However, H. pylori evades the host immune response by inducing Th2 cells and regulatory T cells (Tregs) that produce anti-inflammatory interleukin (IL)-10. Suppression of anti-bacterial Th1/Th17 cells by Tregs may enhance gastric H. pylori propagation, followed by a cascade reaction involving vitamin B12 and folic acid malabsorption, plasma homocysteine elevation, and reactive oxygen species induction. This can damage the blood-brain barrier (BBB), leading to accumulation of amyloid-β in the brain, a hallmark of AD. On the other hand, this suppression of pro-inflammatory Th1/Th17 responses to H. pylori has protective effects on the hosts, since it prevents uncontrolled gastritis as well as suppresses the induction of encephalitogenic Th1/Th17 cells, which can mediate neuroinflammation in MS. The above scenario may explain why chronic H. pylori infection is positively associated with AD, while it is negatively associated with MS. Lastly, we list "10 pitfalls of microbiota studies", which will be useful for evaluating and designing clinical and experimental microbiota studies.
Collapse
Affiliation(s)
- Ah-Mee Park
- Department of Microbiology, Kindai University Faculty of Medicine, 377-2 Ohnohigashi, Osakasayama, Osaka 589-8511, Japan
| | - Seiichi Omura
- Department of Microbiology, Kindai University Faculty of Medicine, 377-2 Ohnohigashi, Osakasayama, Osaka 589-8511, Japan
| | - Mitsugu Fujita
- Department of Microbiology, Kindai University Faculty of Medicine, 377-2 Ohnohigashi, Osakasayama, Osaka 589-8511, Japan
| | - Fumitaka Sato
- Department of Microbiology, Kindai University Faculty of Medicine, 377-2 Ohnohigashi, Osakasayama, Osaka 589-8511, Japan
| | - Ikuo Tsunoda
- Department of Microbiology, Kindai University Faculty of Medicine, 377-2 Ohnohigashi, Osakasayama, Osaka 589-8511, Japan
| |
Collapse
|
444
|
Harris VC, Haak BW, Boele van Hensbroek M, Wiersinga WJ. The Intestinal Microbiome in Infectious Diseases: The Clinical Relevance of a Rapidly Emerging Field. Open Forum Infect Dis 2017; 4:ofx144. [PMID: 28852682 PMCID: PMC5570093 DOI: 10.1093/ofid/ofx144] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 07/06/2017] [Indexed: 02/07/2023] Open
Abstract
The field of infectious disease is undergoing a paradigm shift as the intestinal microbiome is becoming understood. The aim of this review is to inform infectious disease physicians of the potential relevance of the intestinal microbiome to their practice. We searched Medline using both index and text words relating to infectious diseases, microbiome, and probiotics. Relevant articles published up through 2017 were reviewed within Rayyan. The review illustrates pathophysiologic concepts linking the microbiome and infectious diseases; specifically, the intestinal microbiome’s relevance to early immune development, the microbiome and enteric infections, the microbiome’s relevance in compromised hosts, and antimicrobial resistance. Within each subject, there are specific examples of diseases and at-risk patient populations where a role for the microbiome has been strongly established. This provides an overview of the significance of the intestinal microbiome to microbiology, pediatric and adult infectious diseases with an underpinning of concepts useful for the practicing clinician.
Collapse
Affiliation(s)
- Vanessa C Harris
- Department of Medicine, Division of Infectious Diseases and Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, Netherlands.,Amsterdam Institute for Global Health and Development, Amsterdam, Netherlands
| | - Bastiaan W Haak
- Department of Medicine, Division of Infectious Diseases and Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, Netherlands
| | - Michaël Boele van Hensbroek
- Amsterdam Institute for Global Health and Development, Amsterdam, Netherlands.,Emma Children's Hospital, Academic Medical Center, Amsterdam, Netherlands
| | - Willem J Wiersinga
- Department of Medicine, Division of Infectious Diseases and Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, Netherlands
| |
Collapse
|
445
|
Stewart CJ, Embleton ND, Clements E, Luna PN, Smith DP, Fofanova TY, Nelson A, Taylor G, Orr CH, Petrosino JF, Berrington JE, Cummings SP. Cesarean or Vaginal Birth Does Not Impact the Longitudinal Development of the Gut Microbiome in a Cohort of Exclusively Preterm Infants. Front Microbiol 2017. [PMID: 28634475 PMCID: PMC5459931 DOI: 10.3389/fmicb.2017.01008] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The short and long-term impact of birth mode on the developing gut microbiome in neonates has potential implications for the health of infants. In term infants, the microbiome immediately following birth across multiple body sites corresponds to birth mode, with increased Bacteroides in vaginally delivered infants. We aimed to determine the impact of birth mode of the preterm gut microbiome over the first 100 days of life and following neonatal intensive care unit (NICU) discharge. In total, 867 stool samples from 46 preterm infants (21 cesarean and 25 vaginal), median gestational age 27 weeks, were sequenced (V4 region 16S rRNA gene, Illumina MiSeq). Of these, 776 samples passed quality filtering and were included in the analysis. The overall longitudinal alpha-diversity and within infant beta-diversity was comparable between cesarean and vaginally delivered infants. Vaginally delivered infants kept significantly more OTUs from 2 months of life and following NICU discharge, but OTUs lost, gained, and regained were not different based on birth mode. Furthermore, the temporal progression of dominant genera was comparable between birth modes and no significant difference was found for any genera following adjustment for covariates. Lastly, preterm gut community types (PGCTs) showed some moderate differences in very early life, but progressed toward a comparable pattern by week 5. No PGCT was significantly associated with cesarean or vaginal birth. Unlike term infants, birth mode was not significantly associated with changes in microbial diversity, composition, specific taxa, or overall microbial development in preterm infants. This may result from the dominating effects of NICU exposures including the universal use of antibiotics immediately following birth and/or the lack of Bacteroides colonizing preterm infants.
Collapse
Affiliation(s)
- Christopher J Stewart
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, HoustonTX, United States
| | - Nicholas D Embleton
- Newcastle Neonatal ServiceRoyal Victoria Infirmary, Newcastle upon Tyne, United Kingdom
| | - Elizabeth Clements
- School of Science and Engineering, Teesside UniversityMiddlesbrough, United Kingdom
| | - Pamela N Luna
- Department of Statistics, Rice University, HoustonTX, United States
| | - Daniel P Smith
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, HoustonTX, United States
| | - Tatiana Y Fofanova
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, HoustonTX, United States
| | - Andrew Nelson
- Faculty of Health and Life Sciences, Northumbria UniversityNewcastle upon Tyne, United Kingdom
| | - Gillian Taylor
- School of Science and Engineering, Teesside UniversityMiddlesbrough, United Kingdom
| | - Caroline H Orr
- School of Science and Engineering, Teesside UniversityMiddlesbrough, United Kingdom
| | - Joseph F Petrosino
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, HoustonTX, United States
| | - Janet E Berrington
- Newcastle Neonatal ServiceRoyal Victoria Infirmary, Newcastle upon Tyne, United Kingdom
| | - Stephen P Cummings
- School of Science and Engineering, Teesside UniversityMiddlesbrough, United Kingdom
| |
Collapse
|
446
|
Millar M, Seale J, Greenland M, Hardy P, Juszczak E, Wilks M, Panton N, Costeloe K, Wade WG. The Microbiome of Infants Recruited to a Randomised Placebo-controlled Probiotic Trial (PiPS Trial). EBioMedicine 2017; 20:255-262. [PMID: 28571671 PMCID: PMC5478240 DOI: 10.1016/j.ebiom.2017.05.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 05/07/2017] [Accepted: 05/15/2017] [Indexed: 12/17/2022] Open
Abstract
The microbial dysbiosis associated with necrotizing enterocolitis (NEC) in preterm infants suggests that early exposure to probiotics may decrease and antibiotics may increase NEC risk. However, administration of Bifidobacterium breve strain BBG-001 to preterm infants did not affect NEC incidence in a multicenter randomised controlled phase 3 trial (PiPS trial). Using a subset of these subjects we compared the fecal microbiome of probiotic and placebo groups and assessed the impact of early antibiotic treatment. Extracted DNA from 103 fecal samples collected at 36 weeks post-menstrual age underwent PCR amplification of a fragment of the 16S rRNA gene. Heatmaps were constructed showing the proportions of sequences from bacterial families present at > 1% of the community. Stepwise logistic regression assessed the association between early antibiotic exposure and microbiome group. There was no difference in the microbial richness and diversity of the microbiome of preterm infants following treatment with probiotic or a placebo. Conversely, early antimicrobial exposure was associated with different patterns of colonisation, specifically a relative abundance of Proteobacteria. These findings highlight that the potential influence of probiotics on the microbiome of preterm infants remains unclear whereas the modulatory effect of antibiotic exposure on microbial colonisation requires further research. Microbial dysbiosis has been associated with the development of neonatal necrotizing enterocolitis in premature infants. Early administration of a probiotic compared to a placebo did not alter the microbiome of premature infants. Early life exposure to antibiotics was associated with different patterns of colonisation in premature infants.
The association of altered microbial gastrointestinal colonisation (dysbiosis) with neonatal necrotizing enterocolitis suggests that, through their effects on microbial colonisation, probiotics and antimicrobials may modulate necrotizing enterocolitis risk. We compared microbial colonisation patterns of premature infants administered a probiotic or placebo in the early stages of life. We found no significant effect of probiotic administration on microbial colonisation, a finding which disputes the suggestion that prophylactic use of probiotics prevents the dysbiosis associated with necrotizing enterocolitis. In contrast, antibiotic exposure did alter the pattern of colonisation, indicating the need for further research in order to clearly ascertain the nature of this relationship.
Collapse
Affiliation(s)
- Michael Millar
- Department of Infection, Barts Health NHS Trust, London, UK.
| | - Jo Seale
- Department of Infection, Barts Health NHS Trust, London, UK
| | - Melanie Greenland
- National Perinatal Epidemiology Unit, University of Oxford, Oxford, UK
| | - Pollyanna Hardy
- National Perinatal Epidemiology Unit, University of Oxford, Oxford, UK
| | - Edmund Juszczak
- National Perinatal Epidemiology Unit, University of Oxford, Oxford, UK
| | - Mark Wilks
- Department of Infection, Barts Health NHS Trust, London, UK
| | - Nicola Panton
- Department of Infection, Barts Health NHS Trust, London, UK
| | - Kate Costeloe
- Centre for Paediatrics, Blizard Institute, Barts and The London School of Medicine and Dentistry, London, UK
| | - William G Wade
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
447
|
Stewart CJ, Embleton ND, Clements E, Luna PN, Smith DP, Fofanova TY, Nelson A, Taylor G, Orr CH, Petrosino JF, Berrington JE, Cummings SP. Cesarean or Vaginal Birth Does Not Impact the Longitudinal Development of the Gut Microbiome in a Cohort of Exclusively Preterm Infants. Front Microbiol 2017. [PMID: 28634475 DOI: 10.3389/fmicb.2017.01008/full] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023] Open
Abstract
The short and long-term impact of birth mode on the developing gut microbiome in neonates has potential implications for the health of infants. In term infants, the microbiome immediately following birth across multiple body sites corresponds to birth mode, with increased Bacteroides in vaginally delivered infants. We aimed to determine the impact of birth mode of the preterm gut microbiome over the first 100 days of life and following neonatal intensive care unit (NICU) discharge. In total, 867 stool samples from 46 preterm infants (21 cesarean and 25 vaginal), median gestational age 27 weeks, were sequenced (V4 region 16S rRNA gene, Illumina MiSeq). Of these, 776 samples passed quality filtering and were included in the analysis. The overall longitudinal alpha-diversity and within infant beta-diversity was comparable between cesarean and vaginally delivered infants. Vaginally delivered infants kept significantly more OTUs from 2 months of life and following NICU discharge, but OTUs lost, gained, and regained were not different based on birth mode. Furthermore, the temporal progression of dominant genera was comparable between birth modes and no significant difference was found for any genera following adjustment for covariates. Lastly, preterm gut community types (PGCTs) showed some moderate differences in very early life, but progressed toward a comparable pattern by week 5. No PGCT was significantly associated with cesarean or vaginal birth. Unlike term infants, birth mode was not significantly associated with changes in microbial diversity, composition, specific taxa, or overall microbial development in preterm infants. This may result from the dominating effects of NICU exposures including the universal use of antibiotics immediately following birth and/or the lack of Bacteroides colonizing preterm infants.
Collapse
Affiliation(s)
- Christopher J Stewart
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, HoustonTX, United States
| | - Nicholas D Embleton
- Newcastle Neonatal ServiceRoyal Victoria Infirmary, Newcastle upon Tyne, United Kingdom
| | - Elizabeth Clements
- School of Science and Engineering, Teesside UniversityMiddlesbrough, United Kingdom
| | - Pamela N Luna
- Department of Statistics, Rice University, HoustonTX, United States
| | - Daniel P Smith
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, HoustonTX, United States
| | - Tatiana Y Fofanova
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, HoustonTX, United States
| | - Andrew Nelson
- Faculty of Health and Life Sciences, Northumbria UniversityNewcastle upon Tyne, United Kingdom
| | - Gillian Taylor
- School of Science and Engineering, Teesside UniversityMiddlesbrough, United Kingdom
| | - Caroline H Orr
- School of Science and Engineering, Teesside UniversityMiddlesbrough, United Kingdom
| | - Joseph F Petrosino
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, HoustonTX, United States
| | - Janet E Berrington
- Newcastle Neonatal ServiceRoyal Victoria Infirmary, Newcastle upon Tyne, United Kingdom
| | - Stephen P Cummings
- School of Science and Engineering, Teesside UniversityMiddlesbrough, United Kingdom
| |
Collapse
|