Relative abundances of bacterial genera in BF-IAP, BF-C, MF-IAP and MF-C faecal samples. Other category contains genera present at <1% of relative abundance. doi:10.1371/journal.pone.0157527.g003  

Relative abundances of bacterial genera in BF-IAP, BF-C, MF-IAP and MF-C faecal samples. Other category contains genera present at <1% of relative abundance. doi:10.1371/journal.pone.0157527.g003  

Source publication
Article
Full-text available
The faecal microbiota composition of infants born to mothers receiving intrapartum antibiotic prophylaxis with ampicillin against group B Streptococcus was compared with that of control infants, at day 7 and 30 of life. Recruited newborns were both exclusive breastfed and mixed fed, in order to also study the effect of dietary factors on the microb...

Contexts in source publication

Context 1
... infants had significantly higher abundances of Proteobacteria (P<0.062) at day 7 compared to BF-C infants (Fig 2). BF-IAP infants were dominated by genera belonging to the Enterobacteriaceae family (p = 0.044), particularly Escherichia which accounted for 52% of the total relative abundance, compared with 14% in the BF-C group (Fig 3). Bifidobacteria were not detected in any of the BF-IAP infants at day 7; in contrast, Bifidobacterium represented 16% of the relative abundance of the microbiota from BF-C infants (p = 0.001). ...
Context 2
... were not detected in any of the BF-IAP infants at day 7; in contrast, Bifidobacterium represented 16% of the relative abundance of the microbiota from BF-C infants (p = 0.001). BF-C infants also had higher levels of Bacteroides, 20% compared with 7% in BF-IAP, although not statistically significant (p = 0.078) (Fig 3). By day 30, bifido- bacteria numbers appeared to have recovered in the BF-IAP group and now account for 6% of the relative abundance (p = 0.025) in both BF groups. ...
Context 3
... (8%) and Bacteroidetes (36%) were highest in the MF-C group compared with 1% and 21% in MF-IAP infants. MF-IAP infants contained high abundances of organisms belonging to family Enterobacteriaceae, 35%, and Streptococcus, 32% (Fig 3), compared with 17% and 10% in the control group. MF-C infants had higher levels of Bacteroides, 32%, and Bifidobacterium, 5%, compared with 13% and 1%, respectively, in MF-IAP infants (Fig 3). ...
Context 4
... infants contained high abundances of organisms belonging to family Enterobacteriaceae, 35%, and Streptococcus, 32% (Fig 3), compared with 17% and 10% in the control group. MF-C infants had higher levels of Bacteroides, 32%, and Bifidobacterium, 5%, compared with 13% and 1%, respectively, in MF-IAP infants (Fig 3). At day 30, Bacteroidetes were the dominant phylum in both groups, representing 26% in control and 34% in IAP treated infants. ...
Context 5
... Fig 3 shows that Veillonella is affected by the antibiotic treatment, as it does not increase in the BF-IAP group between 7 and 30 days, whereas a strong increase is shown within BF-C samples at the same sampling times (40%) and in BF-C samples with respect to BF-IAP at 30 days (43%). ...
Context 6
... infants were also found to have higher Veillonella, 22%, compared with 4% in the MF-IAP group. Streptococcus were abundant in the MF-IAP infants 32% versus 12% in BF-IAP and Bacteroides were also highest in the MF-IAP group, 13%, compared with 6% in BF-IAP (Fig 3). ...

Similar publications

Article
Full-text available
The natural ingredients' potential as immunostimulants is a plant Curcuma and Aromatic ginger. This research was conducted from March to June 2022 in the Laboratory of Parasites and Fish Diseases, Faculty of Fisheries and Marine, Universitas Riau. Histological preparations were carried out at the Bukittinggi Veterinary Center (BVet). This research...
Article
Full-text available
Brain abscess is a focal intracerebral infection consisting of an encapsulated collection of pus caused by various agents such as bacteria, mycobacteria, fungi, protozoa, or helminths. The incidence is 0.3-1.3 per 100,000 people/year; it may be higher in certain risk groups, including patients with HIV/AIDS. The most frequent locations are: frontal...
Article
Full-text available
Streptococcus agalactiae is an invasive multi-host pathogen that causes invasive diseases mainly in newborns, elderly, and individuals with underlying health complications. In fish, S. agalactiae causes streptococcosis, which is characterized by septicemia and neurological signs, and leads to great economic losses to the fish farming industry world...
Article
Full-text available
The study was conducted in order to evaluate the antibacterial potential of acacia leaves, barks and roots aqueous extracts against the bacterium S. agalactiae by measuring the zone of inhibition and determining the minimum inhibitory concentration. Extract concentrations that recorded at least 50% survival on the bioassay of Nile tilapia fingerlin...

Citations

... Core microbiome genera are widely distributed and abundant across samples. Consistent with prior research [28][29][30], we found that exposure to IAP reduced the abundance of the core genus Bifidobacterium in infants within three days of age. Conversely, in healthy full-term infants without IAP exposure, Bifidobacterium dominated the gut. ...
Article
Full-text available
We aimed to examine the effects of antibiotic and probiotic usage on the gut microbiota structure and the presence of antibiotic-resistance genes (ARGs) in infants during the first six months of life. Questionnaires and fecal samples were collected within three days of birth, two months, and six months to assess antibiotic and probiotic exposure. Gut microbiotas were sequenced via 16S rRNA, and ARGs were conducted by qPCR, including beta-lactam (mecA, blaTEM), tetracycline (tetM), fluoroquinolone (qnrS), aminoglycoside (aac(6′)-Ib), and macrolide (ermB). Infants were categorized by antibiotic and probiotic usage and stratified by delivery mode, microbial composition, and ARG abundances were compared, and potential correlations were explored. A total of 189 fecal samples were analyzed in this study. The gut microbiota diversity (Chao1 index) was significantly lower in the “only probiotics” (PRO) group compared to the “neither antibiotics nor probiotics” (CON) group at six months for the CS stratification (p = 0.029). Compositionally, the abundance of core genus Bifidobacterium_pseudocatenulatum was less abundant for the antibiotic during delivery (IAP) group than that in the CON group within the first three days (p = 0.009), while core genus Enterococcus_faecium was more abundant in the PRO than that in the CON group (p = 0.021) at two months. ARGs were highly detected, with Enterococcus hosting tetM and Escherichia associated with blaTEM within three days of birth, though no correlation was found between Bifidobacterium and ARGs. These findings emphasized the critical importance of carefully managing antibiotic and probiotic exposures in early life, with implications for promoting lifelong health through preserving a healthy infant gut ecosystem.
... 73 It has also been previously described that intrapartum antibiotic prophylaxis (IAP) has a negative effect on the later bifidobacterial establishment in the neonatal gut. 7,74 To assess this effect, antibiotic exposure of our cohort was recorded during pregnancy and also during the first month of life of the infants. During a c-section procedure, antibiotics are commonly administered as a preventive measure to reduce the risk of infection (e.g., cefazolin, vancomycin, clindamycin and erythromycin). ...
Article
Full-text available
Resistance to antibiotics in newborns is a huge concern as their immune system is still developing, and infections and resistance acquisition in early life have short- and long-term consequences for their health. Bifidobacterium species are important commensals capable of dominating the infant gut microbiome and are known to be less prone to possess antimicrobial resistance genes than other taxa that may colonize infants. We aimed to study the association between Bifidobacterium-dominated infant gut microbiota and the antibiotic resistant gene load in neonates, and to ascertain the perinatal factors that may contribute to the antibiotic resistance acquisition. Two hundred infant fecal samples at 7 days and 1 month of age from the MAMI birth cohort were included in the study and for whom maternal-neonatal clinical records were available. Microbiota profiling was carried out by 16S rRNA amplicon sequencing, and targeted antibiotic resistance genes (ARGs) including tetM, tetW, tetO, blaTEM, blaSHV and ermB were quantified by qPCR. Infant microbiota clustered into two distinct groups according to their Bifidobacterium genus abundance: high and low. The main separation of groups or clusters at each time point was performed with an unsupervised non-linear algorithm of k-means partitioning to cluster data by time points based on Bifidobacterium genus relative abundance. Microbiota composition differed significantly between both groups, and specific bifidobacterial species were enriched in each cluster. Lower abundance of Bifidobacterium in the infant gut was associated with a higher load of antibiotic resistance genes. Our results highlight the relevance of Bifidobacterium genus in the early acquisition and establishment of antibiotic resistance in the gut. Further studies are needed to develop strategies to promote a healthy early colonization and fight against the spread of antibiotic resistances.
... Studies focusing entirely on VD neonates of mothers positive for GBS screening present similar ndings. Mazzola et al. (77) found differences within the Enterobacteriaceae family and within the Bi dobacterium and Bacteroides genera in stool samples 7 days after birth. By qPCR quanti cation of selected bacteria, Aloisio et al. (30) and Corvaglia et al. (75) also showed a signi cant average reduction in the counts of Bi dobacterium spp. in neonates whose mothers received IAP. ...
Preprint
Full-text available
Background:Intrapartum antibiotic prophylaxis (IAP) is commonly used during C-section delivery and in Group B Streptococcus-positive women before vaginal delivery. Here, we primarily aimed to investigate the effect of IAP on the neonatal oral and fecal bacteriomes in the first week of life. Methods: In this preliminary study, maternal and neonatal oral swabs and neonatal fecal (meconium and transitional stool) swabs were selected from a pool of samples from healthy mother-neonate pairs participating in the pilot phase of CELSPAC: TNG during their hospital stay. The DNA was extracted and bacteriome profiles were determined by 16S rRNA amplicon sequencing (Illumina). Results: In the final dataset, 33 mother-neonate pairs were exposed to antibiotics during C-section‑ or vaginal delivery (cases; +IAP) and the vaginal delivery without IAP (controls, -IAP) took place in 33 mother-neonate pairs. Alpha diversity (Shannon index, p=0.01), bacterial composition (PERMANOVA, p<0.05), and relative abundance of the genus Gemella (q=0.08) only in neonatal oral samples collected ≤48 h after birth were lower in +IAP than in -IAP group. No significant differences between meconium bacteriomes of the +IAP and -IAP groups were observed (p>0.05). However, the IAP was associated with decreased alpha diversity (number of amplicon sequence variants, p<0.001), decreased relative abundances of the genera Bacteroidesand Bifidobacteria, and increased relative abundances of genera Enterococcusand Rothia (q<0.01 for all of them) in transitional stool samples. Conclusion: The findings of this study suggest that exposure to IAP may significantly influence the early development of the neonatal oral and gut microbiomes. IAP affected the neonatal oral bacteriome in the first two days after birth as well as the neonatal fecal bacteriome in transitional stool samples. In addition, it highlights the necessity for further investigation into the potential long-term health impacts on children.
... Если беременная вынужденно получает антибактериальную терапию, это негативно отражается на составе и ее микробиоты, и микробиоты ребенка после вагинальных родов [14][15][16]. В систематическом обзоре проанализировано 24 исследования: в большинстве публикаций отмечено снижение микробного разнообразия в кишечной микробиоте у детей, рожденных женщинами, получавшими антибактериальную терапию во время родов, в сравнении с младенцами матерей, не получавших такого лечения [14]. Помимо этого, влияние интранатального воздействия антибиотиков проявилось в снижении в микробиоте уровней бактероидов и бифидобактерий при увеличении количества протеобактерий, причем наиболее отчетливо указанные изменения проявились у доношенных детей при вагинальных родах [14]. ...
... Помимо этого, влияние интранатального воздействия антибиотиков проявилось в снижении в микробиоте уровней бактероидов и бифидобактерий при увеличении количества протеобактерий, причем наиболее отчетливо указанные изменения проявились у доношенных детей при вагинальных родах [14]. В сравнительном когортном исследовании оценивали кишечную микробиоту у детей, рожденных матерями, получившими интранатально ампициллин с целью профилактики стрептококковой инфекции, и у детей контрольной группы [15]. Наиболее заметные отличия состава микробиоты были отмечены у новорожденных на грудном вскармливании, они заключались в более высоком относительном содержании Enterobacteriaceae и в более низком бактериальном разнообразии в сравнении с детьми контрольной группы и младенцами на смешанном вскармливании. ...
... Наиболее заметные отличия состава микробиоты были отмечены у новорожденных на грудном вскармливании, они заключались в более высоком относительном содержании Enterobacteriaceae и в более низком бактериальном разнообразии в сравнении с детьми контрольной группы и младенцами на смешанном вскармливании. Уровень бифидобактерий был снижен у всех младенцев у матерей, получивших антибиотик, независимо от характера вскармливания, но к 30-му дню жизни популяция бифидобактерий у этих детей восстанавливалась [15]. Еще в одном систематическом обзоре, включившем 17 обсервационных и 13 рандомизированных контролируемых исследований, указано на противоречивые результаты работ, посвященных влиянию антибактериальной терапии в родах [16]. ...
Article
This review summarizes stages of intestinal microbiota development in infant and immune responses modulation associated to these stages. The leading role of breastfeeding in the optimal microbiota and associated immune responses development during the first half of child’s life is presented. The biological feasibility of supplemental feeding implementation at the second window of opportunity (4–6 months) is justified, as well as role of supplementation products (including cereal) in adult microbiota development.
... IAP carries some risks for the mother and baby, including anaphylaxis, increased medicalization of labor and neonatal period, increased antibiotic resistance, intestinal complaints, nausea, changes in neonatal bowel microbiome, higher risk of necrotizing enterocolitis and Candida spp. colonization which are theoretically linked to a number of late effects in the child such as allergy, obesity and diabetes [1, [13][14][15][16][17]. ...
Article
Full-text available
Objectives The aim of this study was to evaluate if screening Group B Streptococcus colonization by intrapartum polymerase chain reaction could improve intrapartum administration of antibiotic prophylaxis, compared with antepartum culture screening and analyze the sensitivity and specificity of polymerase chain reaction test. Methods 198 pregnant women with Group B Streptococcus colonization antepartum culture screening were included. When they arrived at hospital for delivery, two rectovaginal swabs were collected: for culture and polymerase chain reaction method. Results The rate of Group B Streptococcus colonization antepartum detected by culture was 16.7%; at delivery was 17.2% when detected by culture and 19.7% using polymerase chain reaction method. The rate of inconclusive polymerase chain reaction tests was 0.5%. Considering intrapartum culture screening as gold standard, sensitivity and specificity of polymerase chain reaction test for intrapartum Group B Streptococcus colonization was 97.1% and 95.7%, respectively. The global rate of discordance between antepartum and intrapartum Group B Streptococcus colonization was 6.6%. The rate of women not treated with intrapartum antibiotic prophylaxis in the setting of positive intrapartum culture was significantly lower using intrapartum polymerase chain reaction test (0.5%) than with antepartum culture method (3.5%, p = 0.035). Conclusion The use of intrapartum antibiotic prophylaxis can be more efficient when screening Group B Streptococcus colonization intrapartum by polymerase chain reaction test. Polymerase chain reaction method had a good performance in our study, with high sensitivity and specificity.
... The growing body of knowledge on the impact of perinatal antibiotic exposure on the neonatal microbiome is adding a new aspect to the discussion of antibiotic use in pregnancy and during early life. Apart from the proven benefits of prophylactic (e.g., for operative birth or group B streptococci) and therapeutic antibiotics (e.g., for chorioamnionitis or urinary tract infection) during pregnancy and childbirth [3,11,12,13,14], it has been shown that exposure to antibiotics in this critical phase can change the offspring's developing microbiota significantly [15,16]. The initial colonization of the infant's gut at the beginning of life is influenced by several known factors like birth mode, breast-feeding versus formula feeding, microbial transfer by the mother, environment and early life antibiotic exposure [17]. ...
Article
Full-text available
Introduction Antibiotics are powerful drugs to prevent and treat perinatal infections. Overuse of antibiotics leads to antibiotic resistance, has potential side effects and influences the maternal and neonatal microbiome. Patients and Methods We performed a prospective observational study on the prevalence, indications, and prescribing patterns of antibiotics during pregnancy and childbirth. We included women who had given birth after 23+0 weeks of gestation at a single tertiary center in Germany from January 2020 to March 2021. Descriptive statistics and binomial regression were performed to analyze the factors influencing the prescription of antibiotics. Results We included 522 postpartum women into our study. 337 (64.6%) were exposed to antibiotics during pregnancy and/or childbirth. 115 women received antibiotics during pregnancy, 291 during birth. Most antibiotics during pregnancy were prescribed for urinary tract infections (UTIs) (56.0%). Most prescriptions were issued by obstetrics and gynecology physicians (65.8%), followed by hospitals (16.7%) and family medicine physicians (8.8%). Most antibiotics during childbirth were given for a cesarean section (64.3%), followed by preterm rupture of membranes (41.2%). 95.3% of women who had a preterm birth were exposed to antibiotics. In logistic regression models, lower gestational age at birth, higher maternal body-mass-index and smoking were independently associated with antibiotic use during pregnancy and childbirth. Conclusion We found a high rate of antibiotic exposure during pregnancy and childbirth. Our results imply an urgent need for antibiotic stewardship programs in perinatal medicine as well as further research on the effects of perinatal antibiotic exposure on microbiome development and childhood health.
... GBS infections are conventionally treated with antibiotics, but treatment is often complicated by recur-rence or by antibiotic allergy. Several studies have also shown that antibiotic prophylaxis in pregnant women affects the fetal microbiome Mazzola et al., 2016;Qu et al., 2021). ...
Article
Streptococcus agalactiae (Group B Streptococcus, GBS) is primarily known as a major neonatal pathogen. In adults, these bacteria often colonize the gastrointestinal and urogenital tracts. Treatment of infections using antibiotics is often complicated by recurrences caused by multi-resistant streptococci. Endolysin EN534 from prophage A2 of human isolate Streptococcus agalactiae KMB-534 has a modular structure consisting of two terminal catalytic domains, amidase_3 and CHAP, and one central binding domain, LysM. The EN534 gene was cloned into an expression vector, and the corresponding recombinant protein EN534-C was expressed in Escherichia coli in a soluble form and isolated by affinity chromatography. The lytic activity of this endolysin was tested on cell wall substrates from different GBS serotypes, B. subtilis, L. jensenii, and E. coli. The enzyme lysed streptococci, but not beneficial vaginal lactobacilli. The isolated protein is stable in a temperature range of 20 °C to 37 °C. Calcium ions enhanced the activity of the enzyme in the pH range from 5.0 to 8.0. The exolytic activity of EN534-C was observed by time-lapse fluorescence microscopy on a S. agalactiae CCM 6187 substrate. Recombinant endolysin EN534-C may have the potential to become an antimicrobial agent for the treatment of S. agalactiae infections.
... In 2020, a prospective study was carried out to evaluate the impact of the different specific classes of antibiotics administered as IAP. Lower abundance of Bacteroides, Bifidobacterium Ruminococcus, Blautia, and Roseburia and higher proportions of Oscillospora and Veillonella were found at both six weeks and one year of age after the exposure to any class of antibiotics [24] . In addition, the authors concluded that IAP alters the natural development and trajectory of the infant gut microbiome; its effects persist after one year of life, and particular alterations were associated with specific antibiotics. ...
... Cephalosporin entailed a smaller increment in Bifidobacterium and Enterococcus, and when a mix of antibiotics was used, a decrease in E. coli abundance was observed, in comparison with a non-IAP group of neonates. Moreover, differentially abundant functional metagenomes were also observed at one year of age [24] . ...
Article
Full-text available
The perinatal period sets the basis for the later physiological and immune homeostasis of the individual, with the intestinal microbiota being an important contributor to driving this homeostasis development. Therefore, the initial establishment and later development of the microbiota during early life may play a key role in later health. This early establishment of the intestinal microbiota is known to be affected by several factors, with gestational age, delivery mode, and feeding habits being extensively studied ones. Other factors are not so well understood, although knowledge has been accumulating in the last years. Among them, a factor of great relevance is the effect of perinatal exposure to antibiotics. Administration of intrapartum antimicrobial prophylaxis (IAP) to women during the delivery process represents the most common form of exposure to antibiotics during the perinatal period, present in around 30% of deliveries. During the last decade, evidence has accumulated demonstrating that IAP alters intestinal microbiota development in neonates. Moreover, recent evidence indicates that this practice may also be altering the infant intestinal resistome by increasing the levels of some antibiotic resistance genes. This evidence, as reviewed in this manuscript, suggests the interest in promoting the rational use of IAP. This practice has significantly reduced the risk of neonatal infections, but now the accumulating knowledge suggests the need for strategies to minimize its impact on the neonatal microbiota establishment.
... Similar to our results, Tapiainen et al. [8] reported changes in infant gut microbiota from both IAP exposure and IV antibiotics that were still observed at 6 months of age, including the enrichment of Clostridium and the depletion of Bacteroides species. Consistent with what others have reported [8,[23][24][25][26], we found a reduction of Bifidobacteriaceae and Bacteroidaceae in gut microbiota following antibiotic exposure, as well as an increase in Clostridaceae, Lachnospiraceae, Veillonellaceae, and Enterobacteriaceae at both 3-4 months of age and 12 months of age. Colonization of more Proteobacteria (phylum to which Enterobacteriaceae belongs) may be a signal for gut dysbiosis and inflammation [27], while a reduction in important gut microbes may provide room for C. difficile colonization and overgrowth. ...
Article
Full-text available
The relationship between antibiotic use and Clostridioides difficile (C. difficile) has been well established in adults and older children but remains unclear and is yet to be fully examined in infant populations. This study aimed to determine the separate and cumulative impact from antibiotics and household cleaning products on C. difficile colonization in infants. This study included 1429 infants at 3–4 months of age and 1728 infants at 12 months of age from the Canadian Healthy Infant Longitudinal Development (CHILD) birth cohort. The levels of infant antimicrobial exposure were obtained from hospital birth charts and standardized questionnaires. Infant gut microbiota was characterized by Illumina 16S ribosomal ribonucleic acid (rRNA) gene sequencing. Analysis of C. difficile was performed using a quantitative polymerase chain reaction (qPCR). Overall, C. difficile colonized 31% and 46% of infants at 3–4 months and 12 months, respectively. At 3–4 months, C. difficile colonization was significantly higher in infants exposed to both antibiotics and higher (above average) usage of household cleaning products (adjusted odds ratio (aOR) 1.50, 95% CI 1.03–2.17; p = 0.032) than in infants who had the least antimicrobial exposure. This higher colonization persisted up to 12 months of age. Our study suggests that cumulative exposure to systemic antibiotics and higher usage of household cleaning products facilitates C. difficile colonization in infants. Further research is needed to understand the future health impacts.
... Changes in the relative abundance of bacterial taxa associated with IAPexposure included increased Proteobacteria (a phylum that includes several enteric pathogens) [74], Escherichia spp. [75] and Enterobacteriaceae [77] at 1-3 months of age in three countries. IAP-exposure was also associated at age 3 months with decreased Bifidobacterium spp. ...
... IAP-exposure was also associated at age 3 months with decreased Bifidobacterium spp. [75,77] in two cohorts, and an increase in the Firmicutes phylum [78]. Longer term changes included a reduction in the Bacteroidetes phylum [74,76] and Bifidobacterium spp. at 1 year of age [74]. ...
... and facilitate cross-feeding by other species [81]. Breastfeeding is associated with quicker recovery of microbiome α-diversity in IAP-exposed infants in one observational cohort [77]. The benefits of breastfeeding may also extend beyond its prebiotic content [146]. ...
Article
Full-text available
Antibiotics have become a mainstay of healthcare in the past century due to their activity against pathogens. This manuscript reviews the impact of antibiotic use on the intestinal microbiota in the context of mass drug administration (MDA). The importance of the gut microbiota to human metabolism and physiology is now well established, and antibiotic exposure may impact host health via collateral effects on the microbiota and its functions. To gain further insight into how gut microbiota respond to antibiotic perturbation and the implications for public health, factors that influence the impact of antibiotic exposure on the microbiota, potential health outcomes of antibiotic-induced microbiota alterations, and strategies that have the potential to ameliorate these wider antibiotic-associated microbiota perturbations are also reviewed. Graphical Abstract