Antibiotics at birth and later antibiotic courses: effects on gut microbiota

Sofia Ainonen, Mysore V Tejesvi, Md Rayhan Mahmud, Niko Paalanne, Tytti Pokka, Weizhong Li, Karen E Nelson, Jarmo Salo, Marjo Renko, Petri Vänni, Anna Maria Pirttilä, Terhi Tapiainen, Sofia Ainonen, Mysore V Tejesvi, Md Rayhan Mahmud, Niko Paalanne, Tytti Pokka, Weizhong Li, Karen E Nelson, Jarmo Salo, Marjo Renko, Petri Vänni, Anna Maria Pirttilä, Terhi Tapiainen

Abstract

Background: Intrapartum antibiotic prophylaxis (IAP) is widely used, but the evidence of the long-term effects on the gut microbiota and subsequent health of children is limited. Here, we compared the impacts of perinatal antibiotic exposure and later courses of antibiotic courses on gut microbiota.

Methods: This was a prospective, controlled cohort study among 100 vaginally delivered infants with different perinatal antibiotic exposures: control (27), IAP (27), postnatal antibiotics (24), and IAP and postnatal antibiotics (22). At 1 year of age, we performed next-generation sequencing of the bacterial 16S ribosomal RNA gene of fecal samples.

Results: Exposure to the perinatal antibiotics had a clear impact on the gut microbiota. The abundance of the Bacteroidetes phylum was significantly higher in the control group, whereas the relative abundance of Escherichia coli was significantly lower in the control group. The impact of the perinatal antibiotics on the gut microbiota composition was greater than exposure to later courses of antibiotics (28% of participants).

Conclusions: Perinatal antibiotic exposure had a marked impact on the gut microbiota at the age of 1 year. The timing of the antibiotic exposure appears to be the critical factor for the changes observed in the gut microbiota.

Impact: Infants are commonly exposed to IAP and postnatal antibiotics, and later to courses of antibiotics during the first year of life. Perinatal antibiotics have been associated with an altered gut microbiota during the first months of life, whereas the evidence regarding the long-term impact is more limited. Perinatal antibiotic exposure had a marked impact on the infant's gut microbiota at 1 year of age. Impact of the perinatal antibiotics on the gut microbiota composition was greater than that of the later courses of antibiotics at the age of 1 year.

Conflict of interest statement

The authors declare no competing interests.

© 2021. The Author(s).

Figures

Fig. 1. Study design.
Fig. 1. Study design.
IAP refers to intrapartum antibiotic prophylaxis. a Flowchart and comparisons at 1 year of age according to the perinatal antibiotic exposure. b Flowchart and comparisons according to exposure to courses of antibiotics after the perinatal period.
Fig. 2. Relative abundances of the main…
Fig. 2. Relative abundances of the main bacterial phyla and genera of the gut microbiota at 1 year of age.
The gut microbiota of the vaginally delivered term infants at 1 year, according to a perinatal antibiotic exposure and b antibiotic courses for common childhood infections. P values indicate post hoc comparisons (control vs. the various antibiotic groups).
Fig. 3. Krona charts showing the gut…
Fig. 3. Krona charts showing the gut microbiota at one year of age.
a After perinatal antibiotic exposure. b After courses of antibiotics. Summed results for the group in question and the control group.

References

    1. Chai G, et al. Trends of outpatient prescription drug utilization in US children, 2002–2010. Pediatrics. 2012;130:23–31.
    1. Persaud RR, et al. Perinatal antibiotic exposure of neonates in Canada and associated risk factors: a population-based study. J. Matern. Fetal Neonatal Med. 2015;28:1190–1195.
    1. Le Doare K, Heath PT. An overview of global GBS epidemiology. Vaccine. 2013;31:D7–D12.
    1. Moore MR, Schrag SJ, Schuchat A. Effects of intrapartum antimicrobial prophylaxis for prevention of group-B-streptococcal disease on the incidence and ecology of early-onset neonatal sepsis. Lancet Infect. Dis. 2003;3:201–213.
    1. Le Doare K, et al. Intrapartum antibiotic chemoprophylaxis policies for the prevention of group B streptococcal disease worldwide: systematic review. Clin. Infect. Dis. 2017;65:S143–S151.
    1. Schrag SJ, et al. A population-based comparison of strategies to prevent early-onset group B streptococcal disease in neonates. N. Engl. J. Med. 2002;347:233–239.
    1. Van Dyke MK, et al. Evaluation of universal antenatal screening for group B streptococcus. N. Engl. J. Med. 2009;360:2626–2636.
    1. Fjalstad JW, et al. Early-onset sepsis and antibiotic exposure in term infants: a nationwide population based study in Norway. Pediatr. Infect. Dis. J. 2016;35:1–6.
    1. Zimmermann P, Curtis N. Effect of intrapartum antibiotics on the intestinal microbiota of infants: a systematic review. Arch. Dis. Child Fetal Neonatal Ed. 2020;105:201–208.
    1. Azad MB, et al. Impact of maternal intrapartum antibiotics, method of birth and breastfeeding on gut microbiota during the first year of life: a prospective cohort study. BJOG. 2016;123:983–993.
    1. Tapiainen T, et al. Impact of intrapartum and postnatal antibiotics on the gut microbiome and emergence of antimicrobial resistance in infants. Sci. Rep. 2019;9:10635.
    1. Rossignoli A, Clavenna A, Bonati M. Antibiotic prescription and prevalence rate in the outpatient paediatric population: analysis of surveys published during 2000–2005. Eur. J. Clin. Pharm. 2007;63:1099–1106.
    1. Vaz LE, et al. Recent trends in outpatient antibiotic use in children. Pediatrics. 2014;133:375–385.
    1. Murk W, Risnes KR, Bracken MB. Prenatal or early-life exposure to antibiotics and risk of childhood asthma: a systematic review. Pediatrics. 2011;127:1125–1138.
    1. Kronman MP, Zaoutis TE, Haynes K, Feng R, Coffin SE. Antibiotic exposure and IBD development among children: a population-based cohort study. Pediatrics. 2012;130:e794–e803.
    1. Saari A, Virta LJ, Sankilampi U, Dunkel L, Saxen H. Antibiotic exposure in infancy and risk of being overweight in the first 24 months of life. Pediatrics. 2015;135:617–626.
    1. Zven SE, Susi A, Mitre E, Nylund CM. Association between use of multiple classes of antibiotic in infancy and allergic disease in childhood. JAMA Pediatr. 2020;174:199–200.
    1. Chelimo C, Camargo CA, Jr, Morton SMB, Grant CC. Association of repeated antibiotic exposure up to age 4 years with body mass at age 4.5 years. JAMA Netw. Open. 2020;3:e1917577.
    1. Dydensborg Sander S, et al. Association between antibiotics in the first year of life and celiac disease. Gastroenterology. 2019;156:2217–2229.
    1. Korpela K, et al. Intestinal microbiome is related to lifetime antibiotic use in Finnish pre-school children. Nat. Commun. 2016;7:10410.
    1. Edgar RC. UPARSE: highly accurate OTU sequences from microbial amplicon reads. Nat. Methods. 2013;10:996–998.
    1. Schloss PD, et al. Introducing mothur: open-source, platform-independent, community-supported software for describing and comparing microbial communities. Appl. Environ. Microbiol. 2009;75:7537–7541.
    1. McDonald D, et al. The Biological Observation Matrix (BIOM) format or: how I learned to stop worrying and love the ome-ome. Gigascience. 2012;12:7.
    1. Quast C, et al. The SILVA ribosomal RNA gene database project: improved data processing and web-based tools. Nucleic Acids Res. 2013;41:D590–D596.
    1. Bolyen E, et al. Reproducible, interactive, scalable and extensible microbiome data science using QIIME 2. Nat. Biotechnol. 2019;37:852–857.
    1. Ondov BD, Bergman NH, Phillippy AM. Interactive metagenomic visualization in a Web browser. BMC Bioinform. 2011;12:385–385.
    1. Nogacka A, et al. Impact of intrapartum antimicrobial prophylaxis upon the intestinal microbiota and the prevalence of antibiotic resistance genes in vaginally delivered full-term neonates. Microbiome. 2017;5:93.
    1. Mazzola G, et al. Early gut microbiota perturbations following intrapartum antibiotic prophylaxis to prevent group B streptococcal disease. PLoS ONE. 2016;11:e0157527.
    1. Corvaglia L, et al. Infuence of intrapartum antibiotic prophylaxis for group B streptococcus on gut microbiota in the first month of life. J. Pediatr. Gastroenterol. Nutr. 2016;62:304–308.
    1. Coker MO, et al. Specific class of intrapartum antibiotics relates to maturation of the infant gut microbiota: a prospective cohort study. BJOG. 2020;127:217–227.
    1. Vrieze A, et al. Impact of oral vancomycin on gut microbiota, bile acid metabolism, and insulin sensitivity. J. Hepatol. 2014;60:824–831.
    1. Rashid MU, et al. Determining the long-term effect of antibiotic administration on the human normal intestinal microbiota using culture and pyrosequencing methods. Clin. Infect. Dis. 2015;60:S77–S84.
    1. Stewardson AJ, et al. Collateral damage from oral ciprofloxacin versus nitrofurantoin in outpatients with urinary tract infections: a culture-free analysis of gut microbiota. Clin. Microbiol Infect. 2015;21:344.e1–344e11.
    1. Wei S, et al. Short- and long-term impacts of azithromycin treatment on the gut microbiota in children: a double-blind, randomized, placebo-controlled trial. EBioMedicine. 2018;38:265–272.
    1. Raju SC, et al. Antimicrobial drug use in the first decade of life influences saliva microbiota diversity and composition. Microbiome. 2020;8:121.
    1. Korpela K, et al. Antibiotics in early life associate with specific gut microbiota signatures in a prospective longitudinal infant cohort. Pediatr. Res. 2020;88:438–443.
    1. Seedat F, et al. Universal antenatal screening for group B streptococcus may cause more harm than good. BMJ. 2019;364:I463.
    1. Metz TD, et al. Exposure to group B Streptococcal antibiotic prophylaxis and early childhood body mass index in a vaginal birth cohort. J. Matern. Fetal Neonatal Med. 2019;7:1–6.
    1. Tollanes MC, Moster D, Daltveit AK, Irgens LM. Cesarean section and risk of severe childhood asthma: a population-based cohort study. J. Pediatr. 2008;153:112–116.
    1. Huh SY, et al. Delivery by caesarean section and risk of obesity in preschool age children: a prospective cohort study. Arch. Dis. Child. 2012;97:610–616.
    1. Sevelsted A, Stokholm J, Bønnelykke K, Bisgaard H. Cesarean section and chronic immune disorders. Pediatrics. 2015;135:e92–e98.
    1. Yuan C, et al. Association between cesarean birth and risk of obesity in offspring in childhood, adolescence, and early adulthood. JAMA Pediatr. 2016;170:e162385.
    1. Mitselou N, et al. Cesarean delivery, preterm birth, and risk of food allergy: Nationwide Swedish cohort study of more than 1 million children. J. Allergy Clin. Immunol. 2018;142:1510–1514.e2.
    1. Jakobsson HE, et al. Decreased gut microbiota diversity, delayed Bacteroidetes colonisation and reduced Th1 responses in infants delivered by Caesarean section. Gut. 2014;63:559–566.
    1. Penders J, et al. Factors influencing the composition of the intestinal microbiota in early infancy. Pediatrics. 2006;118:511–521.
    1. Bokulich NA, et al. Antibiotics, birth mode, and diet shape microbiome maturation during early life. Sci. Transl. Med. 2016;8:343ra82.
    1. Freedman AL. Urologic diseases in America Project: urologic diseases in North America project: trends in resource utilization for urinary tract infections in children. J. Urol. 2005;173:949–954.
    1. Magruder M, et al. Gut uropathogen abundance is a risk factor for development of bacteriuria and urinary tract infection. Nat. Commun. 2019;10:5521.
    1. Paalanne N, et al. Intestinal microbiome as a risk factor for urinary tract infections in children. Eur. J. Clin. Microbiol. Infect. Dis. 2018;37:1881–1891.
    1. Michail S, et al. Altered gut microbial energy and metabolism in children with non-alcoholic fatty liver disease. FEMS Microbiol. Ecol. 2014;91:1–9.
    1. Zhu L, et al. Characterization of gut microbiomes in nonalcoholic steatohepatitis (NASH) patients: a connection between endogenous alcohol and NASH. Hepatology. 2013;57:601–609.
    1. Kaakoush NO, et al. Microbial dysbiosis in pediatric patients with Crohn’s disease. J. Clin. Microbiol. 2012;50:3258–3266.

Source: PubMed

3
Abonnieren