Gut pathogen colonization precedes bloodstream infection in the neonatal intensive care unit

Author:

Schwartz Drew J.12345ORCID,Shalon Nitan1,Wardenburg Kate1ORCID,DeVeaux Anna1ORCID,Wallace Meghan A.6,Hall-Moore Carla3,Ndao I. Malick3,Sullivan Janice E.7ORCID,Radmacher Paula7,Escobedo Marilyn8,Burnham Carey-Ann D.346ORCID,Warner Barbara B.3ORCID,Tarr Phillip I.34ORCID,Dantas Gautam13469ORCID

Affiliation:

1. Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA.

2. Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA.

3. Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA.

4. Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA.

5. Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO 63110, USA.

6. Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.

7. Department of Pediatrics, University of Louisville School of Medicine, Norton Children’s Hospital, Louisville, KY 40202, USA.

8. Department of Pediatrics, University of Oklahoma, Oklahoma City, OK 73117, USA.

9. Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA.

Abstract

Bacterial bloodstream infections (BSIs) resulting in late-onset sepsis affect up to half of extremely preterm infants and have substantial morbidity and mortality. Bacterial species associated with BSIs in neonatal intensive care units (NICUs) commonly colonize the preterm infant gut microbiome. Accordingly, we hypothesized that the gut microbiome is a reservoir of BSI-causing pathogenic strains that increase in abundance before BSI onset. We analyzed 550 previously published fecal metagenomes from 115 hospitalized neonates and found that recent ampicillin, gentamicin, or vancomycin exposure was associated with increased abundance of Enterobacteriaceae and Enterococcaceae in infant guts. We then performed shotgun metagenomic sequencing on 462 longitudinal fecal samples from 19 preterm infants (cases) with BSI and 37 non-BSI controls, along with whole-genome sequencing of the BSI isolates. Infants with BSI caused by Enterobacteriaceae were more likely than infants with BSI caused by other organisms to have had ampicillin, gentamicin, or vancomycin exposure in the 10 days before BSI. Relative to controls, gut microbiomes of cases had increased relative abundance of the BSI-causing species and clustered by Bray-Curtis dissimilarity according to BSI pathogen. We demonstrated that 11 of 19 (58%) of gut microbiomes before BSI, and 15 of 19 (79%) of gut microbiomes at any time, harbored the BSI isolate with fewer than 20 genomic substitutions. Last, BSI strains from the Enterobacteriaceae and Enterococcaceae families were detected in multiple infants, indicating BSI-strain transmission. Our findings support future studies to evaluate BSI risk prediction strategies based on gut microbiome abundance in hospitalized preterm infants.

Publisher

American Association for the Advancement of Science (AAAS)

Subject

General Medicine

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3