Enteral broad-spectrum antibiotics antagonize the effect of fecal microbiota transplantation in preterm pigs

Research output: Contribution to journalJournal articleResearchpeer-review

Standard

Enteral broad-spectrum antibiotics antagonize the effect of fecal microbiota transplantation in preterm pigs. / Brunse, Anders; Offersen, Simone Margaard; Mosegaard, Josefine Juliane; Deng, Ling; Damborg, Peter; Nielsen, Dennis Sandris; Sangild, Per Torp; Thymann, Thomas; Nguyen, Duc Ninh.

In: Gut Microbes, Vol. 13, No. 1, 2021, p. 1-16.

Research output: Contribution to journalJournal articleResearchpeer-review

Harvard

Brunse, A, Offersen, SM, Mosegaard, JJ, Deng, L, Damborg, P, Nielsen, DS, Sangild, PT, Thymann, T & Nguyen, DN 2021, 'Enteral broad-spectrum antibiotics antagonize the effect of fecal microbiota transplantation in preterm pigs', Gut Microbes, vol. 13, no. 1, pp. 1-16. https://doi.org/10.1080/19490976.2020.1849997

APA

Brunse, A., Offersen, S. M., Mosegaard, J. J., Deng, L., Damborg, P., Nielsen, D. S., Sangild, P. T., Thymann, T., & Nguyen, D. N. (2021). Enteral broad-spectrum antibiotics antagonize the effect of fecal microbiota transplantation in preterm pigs. Gut Microbes, 13(1), 1-16. https://doi.org/10.1080/19490976.2020.1849997

Vancouver

Brunse A, Offersen SM, Mosegaard JJ, Deng L, Damborg P, Nielsen DS et al. Enteral broad-spectrum antibiotics antagonize the effect of fecal microbiota transplantation in preterm pigs. Gut Microbes. 2021;13(1):1-16. https://doi.org/10.1080/19490976.2020.1849997

Author

Brunse, Anders ; Offersen, Simone Margaard ; Mosegaard, Josefine Juliane ; Deng, Ling ; Damborg, Peter ; Nielsen, Dennis Sandris ; Sangild, Per Torp ; Thymann, Thomas ; Nguyen, Duc Ninh. / Enteral broad-spectrum antibiotics antagonize the effect of fecal microbiota transplantation in preterm pigs. In: Gut Microbes. 2021 ; Vol. 13, No. 1. pp. 1-16.

Bibtex

@article{756ca4ff3656419e8910fa3f28828cef,
title = "Enteral broad-spectrum antibiotics antagonize the effect of fecal microbiota transplantation in preterm pigs",
abstract = "Preterm infants are at risk of multiple morbidities including necrotizing enterocolitis (NEC). Suspected NEC patients receive intravenous antibiotics (AB) to prevent sepsis, although enteral AB is arguably more effective at reducing NEC but is rarely used due to the risk of AB resistance. Fecal microbiota transplantation (FMT) has shown protective effects against NEC in animal experiments, but the interaction between AB and FMT has not been investigated in neonates. We hypothesized that administration of enteral AB followed by rectal FMT would effectively prevent NEC with negligible changes in AB resistance and systemic immunity. Using preterm piglets, we examined host and gut microbiota responses to AB, FMT, or a sequential combination thereof, with emphasis on NEC development. In a saline-controlled experiment, preterm piglets (n = 67) received oro-gastric neomycin (50 mg/kg/d) and amoxicillin-clavulanate (50/12.5 mg/kg/d) (hereafter AB) for four days after cesarean delivery, and were subsequently given rectal FMT from healthy suckling piglet donors. Whereas AB protected the stomach and small intestine, and FMT primarily protected the colon, the sequential combination treatment surprisingly provided no NEC protection. Furthermore, minor changes in the gut microbiota composition were observed in response to either treatment, although AB treatment decreased species diversity and increased AB resistance among coliform bacteria and Enterococci, which were both partly reversed by FMT. Besides, enteral AB treatment suppressed cellular and functional systemic immune development, which was not prevented by subsequent FMT. We discovered an antagonistic relationship between enteral AB and FMT in terms of NEC development. The outcome may depend on choice of AB compounds, FMT composition, doses, treatment duration, and administration routes, but these results challenge the applicability of enteral AB and FMT in preterm infants.",
keywords = "antibiotics, antibiotics resistance, fecal microbiota transplantation, Gut microbiota, immunity, necrotizing enterocolitis, prematurity",
author = "Anders Brunse and Offersen, {Simone Margaard} and Mosegaard, {Josefine Juliane} and Ling Deng and Peter Damborg and Nielsen, {Dennis Sandris} and Sangild, {Per Torp} and Thomas Thymann and Nguyen, {Duc Ninh}",
year = "2021",
doi = "10.1080/19490976.2020.1849997",
language = "English",
volume = "13",
pages = "1--16",
journal = "Gut Microbes",
issn = "1949-0976",
publisher = "Taylor & Francis",
number = "1",

}

RIS

TY - JOUR

T1 - Enteral broad-spectrum antibiotics antagonize the effect of fecal microbiota transplantation in preterm pigs

AU - Brunse, Anders

AU - Offersen, Simone Margaard

AU - Mosegaard, Josefine Juliane

AU - Deng, Ling

AU - Damborg, Peter

AU - Nielsen, Dennis Sandris

AU - Sangild, Per Torp

AU - Thymann, Thomas

AU - Nguyen, Duc Ninh

PY - 2021

Y1 - 2021

N2 - Preterm infants are at risk of multiple morbidities including necrotizing enterocolitis (NEC). Suspected NEC patients receive intravenous antibiotics (AB) to prevent sepsis, although enteral AB is arguably more effective at reducing NEC but is rarely used due to the risk of AB resistance. Fecal microbiota transplantation (FMT) has shown protective effects against NEC in animal experiments, but the interaction between AB and FMT has not been investigated in neonates. We hypothesized that administration of enteral AB followed by rectal FMT would effectively prevent NEC with negligible changes in AB resistance and systemic immunity. Using preterm piglets, we examined host and gut microbiota responses to AB, FMT, or a sequential combination thereof, with emphasis on NEC development. In a saline-controlled experiment, preterm piglets (n = 67) received oro-gastric neomycin (50 mg/kg/d) and amoxicillin-clavulanate (50/12.5 mg/kg/d) (hereafter AB) for four days after cesarean delivery, and were subsequently given rectal FMT from healthy suckling piglet donors. Whereas AB protected the stomach and small intestine, and FMT primarily protected the colon, the sequential combination treatment surprisingly provided no NEC protection. Furthermore, minor changes in the gut microbiota composition were observed in response to either treatment, although AB treatment decreased species diversity and increased AB resistance among coliform bacteria and Enterococci, which were both partly reversed by FMT. Besides, enteral AB treatment suppressed cellular and functional systemic immune development, which was not prevented by subsequent FMT. We discovered an antagonistic relationship between enteral AB and FMT in terms of NEC development. The outcome may depend on choice of AB compounds, FMT composition, doses, treatment duration, and administration routes, but these results challenge the applicability of enteral AB and FMT in preterm infants.

AB - Preterm infants are at risk of multiple morbidities including necrotizing enterocolitis (NEC). Suspected NEC patients receive intravenous antibiotics (AB) to prevent sepsis, although enteral AB is arguably more effective at reducing NEC but is rarely used due to the risk of AB resistance. Fecal microbiota transplantation (FMT) has shown protective effects against NEC in animal experiments, but the interaction between AB and FMT has not been investigated in neonates. We hypothesized that administration of enteral AB followed by rectal FMT would effectively prevent NEC with negligible changes in AB resistance and systemic immunity. Using preterm piglets, we examined host and gut microbiota responses to AB, FMT, or a sequential combination thereof, with emphasis on NEC development. In a saline-controlled experiment, preterm piglets (n = 67) received oro-gastric neomycin (50 mg/kg/d) and amoxicillin-clavulanate (50/12.5 mg/kg/d) (hereafter AB) for four days after cesarean delivery, and were subsequently given rectal FMT from healthy suckling piglet donors. Whereas AB protected the stomach and small intestine, and FMT primarily protected the colon, the sequential combination treatment surprisingly provided no NEC protection. Furthermore, minor changes in the gut microbiota composition were observed in response to either treatment, although AB treatment decreased species diversity and increased AB resistance among coliform bacteria and Enterococci, which were both partly reversed by FMT. Besides, enteral AB treatment suppressed cellular and functional systemic immune development, which was not prevented by subsequent FMT. We discovered an antagonistic relationship between enteral AB and FMT in terms of NEC development. The outcome may depend on choice of AB compounds, FMT composition, doses, treatment duration, and administration routes, but these results challenge the applicability of enteral AB and FMT in preterm infants.

KW - antibiotics

KW - antibiotics resistance

KW - fecal microbiota transplantation

KW - Gut microbiota

KW - immunity

KW - necrotizing enterocolitis

KW - prematurity

U2 - 10.1080/19490976.2020.1849997

DO - 10.1080/19490976.2020.1849997

M3 - Journal article

C2 - 33382952

AN - SCOPUS:85098618590

VL - 13

SP - 1

EP - 16

JO - Gut Microbes

JF - Gut Microbes

SN - 1949-0976

IS - 1

ER -

ID: 254989685