Diversification and Evolution of Vancomycin-Resistant Enterococcus faecium during Intestinal Domination.

TitleDiversification and Evolution of Vancomycin-Resistant Enterococcus faecium during Intestinal Domination.
Publication TypeJournal Article
Year of Publication2019
AuthorsDubin KA, Mathur D, McKenney PT, Taylor BP, Littmann ER, Peled JU, van den Brink MRM, Taur Y, Pamer EG, Xavier JB
JournalInfect Immun
Volume87
Issue7
Date Published2019 07
ISSN1098-5522
KeywordsAnimals, Biological Evolution, DNA Mutational Analysis, DNA, Bacterial, Enterococcus faecium, Feces, Genetic Variation, Gram-Positive Bacterial Infections, Humans, Longitudinal Studies, RNA, Ribosomal, 16S, Vancomycin-Resistant Enterococci
Abstract

Vancomycin-resistant (VRE) is a leading cause of hospital-acquired infections. This is particularly true in immunocompromised patients, where the damage to the microbiota caused by antibiotics can lead to VRE domination of the intestine, increasing a patient's risk for bloodstream infection. In previous studies we observed that the intestinal domination by VRE of patients hospitalized to receive allogeneic bone marrow transplantation can persist for weeks, but little is known about subspecies diversification and evolution during prolonged domination. Here we combined a longitudinal analysis of patient data and experiments to reveal previously unappreciated subspecies dynamics during VRE domination that appeared to be stable from 16S rRNA microbiota analyses. Whole-genome sequencing of isolates obtained from sequential stool samples provided by VRE-dominated patients revealed an unanticipated level of VRE population complexity that evolved over time. In experiments with ampicillin-treated mice colonized with a single CFU, VRE rapidly diversified and expanded into distinct lineages that competed for dominance. Mathematical modeling shows that evolution follows mostly a parabolic fitness landscape, where each new mutation provides diminishing returns and, in the setting of continuous ampicillin treatment, reveals a fitness advantage for mutations in penicillin-binding protein 5 () that increase resistance to ampicillin. Our results reveal the rapid diversification of host-colonizing VRE populations, with implications for epidemiologic tracking of in-hospital VRE transmission and susceptibility to antibiotic treatment.

DOI10.1128/IAI.00102-19
Alternate JournalInfect. Immun.
PubMed ID31010813
PubMed Central IDPMC6589067
Grant ListP30 CA008748 / CA / NCI NIH HHS / United States
R01 AI137269 / AI / NIAID NIH HHS / United States
U01 AI124275 / AI / NIAID NIH HHS / United States