Skip to main content

ORIGINAL RESEARCH article

Front. Pediatr., 16 February 2021
Sec. Neonatology
This article is part of the Research Topic Precision Medicine in Neonates View all 12 articles

Clinical Study of Correlation for the Intestinal and Pharyngeal Microbiota in the Premature Neonates

\nSen Yang,&#x;Sen Yang1,2Lina Qiao,&#x;Lina Qiao1,2Jing ShiJing Shi1Liang XieLiang Xie2Yang LiuYang Liu2Ying Xiong
Ying Xiong1*Hanmin Liu,
Hanmin Liu1,2*
  • 1Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
  • 2Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China

Objective: There are mutual influences between intestine and lung, that propose a concept of the gut-lung axis, but the mechanism is still unclear. Microbial colonization in early life plays an important role in regulating intestinal and lung function. In order to explore the characteristics of early microbiota on the gut-lung axis, we studied the correlation between intestinal and pharyngeal microbiota on day 1 and day 28 after birth in premature neonates.

Methods: Thirteen neonates born at 26–32 weeks gestational age (GA) hospitalized at the neonatal intensive care unit (NICU) of the West China Second Hospital of Sichuan University were enrolled in this study. Stool samples and pharyngeal swabs samples were collected from each neonate on the first day (T1) and the 28th day (T28) after birth. Total bacterial DNA was extracted and sequenced using the Illumina MiSeq Sequencing System based on the V3–V4 hyper-variable regions of the 16S rRNA gene. Based on the sequencing results, the composition of the intestinal and pharyngeal microbiota was compared and analyzed.

Results: At T1, the difference in microbial composition between intestine and pharynx was not statistically significant. The intestinal microbiota was mainly composed of Unidentified Enterobacteriaceae, Ralstonia, Streptococcus, Fusobacterium, Ureaplasma, etc. The pharyngeal microbiota was mainly composed of Ureaplasma, Bacteroides, Fusobacterium, etc. Ureaplasma and Fusobacterium were detected in both intestine and pharynx. At T28, there was a significant difference in microbial composition between intestine and pharynx (p < 0.001). The intestinal microbiota was mainly composed of Unidentified Clostridiales, Klebsiella, Unidentified Enterobacteriaceae, Enterobacter, Streptococcus, etc. Pharyngeal microbiota was mainly composed of Streptococcus, Rothia, etc. Streptococcus was detected in both intestine and pharynx.

Conclusions: The intestine and pharynx of premature neonates have a unique microbial composition, and share some common microbiota. Whether these microbiotas play a role in the mechanism of gut-lung crosstalk needs further study.

Introduction

Intestinal microbiota plays an important role in human health and disease (1). In recent years, numerous studies have shown that infant intestinal microbiota affects the growth and development of children (2, 3), and are associated with neonatal sepsis, neonatal necrotizing enterocolitis (4), childhood obesity (5), asthma, eczema (6), and diabetes (7), hypertension (8), and other diseases in adulthood. It is commonly believed that the uterus is sterile and the colonization of microbiota began after birth. But recent research results suggested that the intestinal microbiota could colonize in neonates before delivery. The concept of sterile uterus has been challenged, but the results are still controversial and worth further study (9, 10). The initial colonization of microbiota in the lung may affect the development of the respiratory system and even lead to disease (11, 12). Chronic respiratory diseases can affect the composition of the intestinal microbiota, and in turn, intestinal microbiota may affected the function of the respiratory system (13, 14). These results suggest that there is a crosstalk between the respiratory system and the intestine, which proposes a concept of the gut-lung axis (15). There are many studies on the gut-lung axis, but the mechanism is still unclear (16). Therefore, it is very crucial to study the microbial characteristics of the intestine and lung for further exploring the mechanism of the gut-lung axis.

The pharynx connects the oral cavity, nasal cavity, lower respiratory tract, and digestive tract. It communicates with the outside world and is exposed to a variety of exogenous and endogenous microbes. Thus, the pharynx is an ecological niche for potentially pathogenic microbe, which may cause local inflammation or lead to lung diseases (17). Previous studies have shown that there was a large amount of overlap between the pharyngeal microbiota and the respiratory microbiota (18), so the pharyngeal microbiota could, to some extent, reflect the characteristics of the respiratory microbiota. Besides, the pharyngeal swab is a fairly accurate method to define the composition of the respiratory microbiota. Therefore, studying the composition and characteristic of pharyngeal microbiota may open a window for exploring respiratory microbiota.

The concept of the gut-lung axis was born out of the observation that different lung diseases can be influenced by intestinal microenvironment changes and vice versa. The microbiota is an important factor responsible for interactions between these two sites in asthma (19). In addition, the intestinal microbiota of patients with severe bacterial pneumonia (20), cystic fibrosis (19), and influenza differs from that of healthy controls (21). Many studies show that early life is the most important period during which microbiota dysbiosis in the intestine may lead to the development of many respiratory diseases, as the intestine microbiota has a significant influence on immune cell maturation and resistance to pathogens (22).

In this study, we took premature neonates with GA <32 weeks as study subjects to analyze intestinal and pharyngeal microbial characteristics and influencing factors. Meanwhile, we studied the correlation between intestinal and pharyngeal microbiota, to provide some evidence for the role of early microbiota in the mechanism of the gut-lung axis.

Patients and Methods

Study Participants

Premature neonates who were delivered and hospitalized in the NICU of West China Second Hospital of Sichuan University from December 2019 to May 2020 were enrolled. Selection criteria were as follows: GA < 32 weeks; single birth; the birth weight of the infant appropriate for gestational age (between the 10–90th percentile of the average weight of the infant of the same gestational age) (23, 24). Exclusion criteria were as follows: infants with congenital malformations; intrauterine growth retardation; neonatal hypoxic-ischemic encephalopathy; immunodeficiency or severe infectious diseases; mother's antibiotic therapy for more than 3 days within 2 weeks before delivery; the neonate using antibiotics after birth more than 3 days within 2 weeks before delivery; the mother or the neonate used probiotics or prebiotics during perinatal period; and the guardian does not agree to participate or withdraws from the study. The following information was obtained from the medical records: duration of ruptured membranes; prenatal antibiotic use; delivery mode; GA; birth weight; gender. The study protocol was approved by the medical ethics committee of the West China Second Hospital of Sichuan University, and written informed consents were obtained from the parents or guardians of the neonates.

Sample Collection Method

Stool samples were collected from baby diapers with sterile test tubes at T1 and T28. Pharyngeal secretions samples were collected with sterile pharyngeal swabs within 30 min after birth and T28. Stool and pharyngeal swabs samples were quickly placed in a −20°C refrigerator after being collected, and stored in a −80°C refrigerator within 24 h until further processing.

DNA Extraction and Amplification

Total genome DNA from samples was extracted using Hexadecyltrimethy Ammonium Bromide (CTAB) method. DNA concentration and purity were detected by a spectrophotometer. According to the concentration, DNA was diluted to 1 ng/μl using sterile water. The bacteria genomic DNA was amplified with the 341F and 806R primers specific for the V3–V4 hypervariable regions of the 16S rDNA gene. All DNAs were amplified by following a protocol described previously (25). Samples were sequenced on an Illumina MiSeq platform according to the manufacturer's recommendations. The sequencing service was provided by Beijing Novogene Genomics Technology Co. Ltd (China).

Data Processing

Paired-end reads from the original DNA fragments are merged by using FLASH (26), which is designed to merge paired-end reads when there are overlaps between reads 1 and reads 2. Paired-end reads were assigned to each sample according to the unique barcodes. Sequences were analyzed using QIIME software package (Quantitative Insights Into Microbial Ecology), and in-house Perl scripts were used to analyze alpha (within samples) and beta (among samples) diversity. First, reads were filtered by QIIME quality filters. Then we use pick_de_novo_otus. py to pick operational taxonomic units (OTUs) by making an OTU table. Sequences with ≥97% similarity were assigned to the same OTUs. We pick representative sequences for each OTU and use the RDP classifier (27) to annotate taxonomic information for each representative sequence. In order to compute Alpha Diversity, we rarify the OTU table and calculate the Shannon index and Chao1 estimator. QIIME calculates weighted unifrac, which are phylogenetic measures of beta diversity. We used weighted uniFrac for Principal Coordinate Analysis (PCoA). LefSe tool was implemented to identify differentially abundant taxa.

Statistical Analysis

Continuous variables were reported as means ± standard deviations, and categorical data were presented as ratios or percentages. Non-parametric Wilcoxon rank-sum test was used to study differences in continuous variables and Fisher's exact tests were used to analyze categorical variables. p < 0.05 was considered statistically significant.

Results

Clinical Characteristics, and Influencing Factors of Microbial Colonization

In this study, 22 infants with GA <32 weeks in the NICU of West China Second Hospital of Sichuan University were included. Based on the exclusion criteria, 9 cases were excluded, and the remaining 13 cases were included in the final analysis (Table 1). At T1, 13 meconium samples were collected and included in the T1s group, and 13 pharyngeal swabs samples were collected and included in the T1y group. At T28, 13 stool samples were collected and included in the T28s group, and 13 pharyngeal swabs samples were collected and included in the T28y group. In total, 52 samples were collected from the enrolled neonates, 16S rRNA was amplified from 34 samples (6 samples in T1s group; 4 samples in T1y group; 13 samples in T28s group; 11 samples in T28y group; Table 2), and 2,944,688 reads were obtained. At T1, maternal antibiotics, GA, delivery mode, and gender had some effect on the microbial colonization of meconium and pharyngeal swabs, but these were not significant (Table 3).

TABLE 1
www.frontiersin.org

Table 1. Clinical characteristics of neonates (N = 13).

TABLE 2
www.frontiersin.org

Table 2. The result of 16S rRNA was amplified from samples (O: microbiota was detected).

TABLE 3
www.frontiersin.org

Table 3. Analysis of influencing factors of intestinal and pharyngeal microbial colonization.

Comparisons Between Intestinal and Pharyngeal Microbiota

At T1, 1,340 OTUs were detected in the intestine, and 2,404 OTUs were detected in the pharynx, among which 179 OTUs were commonly shared. At T28, 536 OTUs were detected in the intestine, and 1,377 OTUs were detected in the pharynx, among which, 120 OTUs were commonly shared (Figure 1A). At T1, there was no significant difference in Shannon index and Chao1 index between the T1s group and the T1y group. At T28 there was no significant difference in Shannon index between the T28s group and the T28y group; the Chao1 index of the T28y group was higher than that of the T28s group, the difference was significant (p < 0.05; Figure 1B).

FIGURE 1
www.frontiersin.org

Figure 1. Comparisons between intestinal and pharyngeal microbiota. (A) OTUs were shared by the intestine and pharynx. (B) Comparison of the microbial biodiversity between intestine and pharynx, the shannon index, and chao1 were shown as estimators. (C) PCoA plot based on OTU abundance. Each point represents the intestinal or pharyngeal microbiota of a subject. (D) Histogram of the LDA scored for differentially abundant genera between intestine and pharynx. LDA scores were calculated by LDA effect size using linear discriminant analysis.

In order to compare the composition of the overall microbiota of the intestine and pharynx, PCoA was implemented based on the OTU level. At T1, the results of PCoA showed no difference in microbiota composition between the T1s group and the T1y group. At T28, the results of PCoA showed a significant difference in microbiota composition between the T28s group and the T28y group (p < 0.001; Figure 1C). Next, the LEfSe tool (28) was used to analyze microbiota in stool samples and pharyngeal swabs samples, and to detect potential significant differences in relative abundances between the intestinal and pharyngeal microbiota. There were significant differences in 2 genera between the T1s group and the T1y group. Ureaplasma was relatively more abundant in the T1y group, whereas Unidentified Enterobacteriaceae was relatively more abundant in the T1s group. There were significant differences in some genera between the T28s group and the T28y group. Streptococcus, Rothia were relatively more abundant in the T28y group, whereas Unidentified Clostridiales, Unidentified Enterobacteriaceae, Klebsiella were relatively more abundant in the T28s group (Figure 1D).

The Microbial Composition of the Intestine and Pharynx at the Genus Level

We analyzed the microbial composition of the intestine and pharynx at the genus level and found that all samples were dominated by a specific genus (Figure 2A). At T1, intestinal microbiota was mainly composed of Unidentified Enterobacteriaceae (18.8%), Ralstonia (16.7%), Streptococcus (16.6%), Fusobacterium (14.2%), Ureaplasma (11.9%), etc. Pharyngeal microbiota was mainly composed of Ureaplasma (38.1%), Bacteroides (14.9%), Fusobacterium (1.8%), etc. At T28, intestinal microbiota was mainly composed of Unidentified Clostridiales (30.0%), Klebsiella (19.6%), Unidentified Enterobacteriaceae (18.8%), Enterobacter (7.0%), Streptococcus (2.0%), etc. Pharyngeal microbiota was mainly composed of Streptococcus (70.7%), Rothia (17.6%), etc. At T1, the relative abundance of Ureaplasma between the T1s group and the T1y group was 11.9 vs. 38.1% (p = 0.053); The relative abundance of Unidentified Enterobacteriaceae between the T1s group and the T1y group was 18.4 vs. 0.08% (p < 0.05). At T28, the relative abundance of Streptococcus between the T28s group and the T28y group was 2.0 vs. 70.7% (p < 0.05); the relative abundance of Rothia between the T28s group and the T28y group was 0.1 vs. 17.6% (p < 0.05); the relative abundance of Unidentified Clostridiales between the T28s group and the T28y group was 30.0 vs. 0.002% (p < 0.05); the relative abundance of Unidentified Enterobacteriaceae between the T28s group and the T28y group was 18.8 vs. 0.02% (p < 0.05); the relative abundance of Klebsiella between the T28s group and the T28y group was 19.6 vs. 0.5% (p < 0.05; Figure 2B).

FIGURE 2
www.frontiersin.org

Figure 2. The composition of intestinal and pharyngeal microbiota.at the genus level. (A) The microbial composition of each sample. (B) Average microbiota of T1s, T1y, T28s, and T28y (T1s: meconium samples at T1; T1y: pharyngeal swabs samples at T1; T28s: stool samples at T28; T28y: pharyngeal swabs samples at T28).

Discussion

Microbial colonization is a complex and dynamic process. In this study, premature neonates with GA <32 weeks were selected as study subjects. Microbiota was detected in 6 meconium samples and 4 pharyngeal swabs samples at birth, which seems to be a challenge to the concept of a sterile uterus, but it needs to be determined by a larger sample size and the exclusion of more interfering factors. Maternal antibiotics, GA, delivery mode, and gender had no significant effect on the microbial detection rate of meconium and pharyngeal swabs, this is inconsistent with the results of some previous studies that the delivery mode affects the microbiota colonization of the neonates (29). The possible reason is that our sample collection was carried out immediately after birth, and the colonization of environmental microbiota has not been completed.

Through Venn diagram analysis, at T1, intestine and pharynx shared more OTUs than at T2. We may speculate that this pattern of colonization can be driven by the developing host immunity (30), or, alternatively, one pioneer group of bacteria (31) might metabolically prime the environment for replacement by a particular successor. Our observations support the hypothesis that temporal factors influence the microbial colonization of preterm infants, such as there was no significant difference in Shannon index between the T28s group and the T28y group. The Chao1 index of the T28y group was higher than that of the T28s group, and the difference was significant. The results show that the pharyngeal microbiota diversity was higher than that of the intestinal and that the low abundance and rare species are present in the community at T28y group. Through PCoA analysis, there was no significant difference in microbial composition between the T1s group and the T1y group. At T28, there was a significant difference of microbial composition between the T28s group and the T28y group. Streptococcus, Rothia were relatively more abundant in the T28y group, whereas Unidentified Clostridiales, Klebsiella, Unidentified Enterobacteriaceae, Enterobacter were relatively more abundant in the T28s group.

The analysis of microbial composition show that, at T1, the intestine was dominated by Unidentified Enterobacteriaceae, Ralstonia, Streptococcus, etc. among which Unidentified Enterobacteriaceae and Streptococcus are potential pathogenic microbes, and the rest are mostly pathogenic microbes. It suggests that premature neonates have very few beneficial microbes in the intestine, but a large number of potentially pathogenic microbes and pathogenic microbes, which is consistent with previous studies (32). At T1, pharyngeal microbiota was mainly composed of Ureaplasma, Bacteroides, Fusobacterium, etc, which are similar to the oropharyngeal microbiota of infants (33, 34). Ureaplasma was detected in both intestine and pharynx, the relative abundance has no significant difference between the T1s group and the T1y group. This may indicate that the origin of intestinal and pharyngeal microbiota may be the same at birth. The relative abundance and composition of the intestinal and pharyngeal microbiota was significantly different on the 28th day after birth. Streptococcus mainly existed in the pharynx, while Unidentified Clostridiales mainly existed in the intestine. We analyzed the characteristics of intestinal microbiota at different time, the results show that the intestinal microbiota colonization with beneficial bacteria is delayed in preterm infants, while the number of potentially pathogenic bacteria is high. Ureaplasma was relatively more abundant in the T1y group, Streptococcus was relatively more abundant in the T28y group. There was significant difference between early and late pharyngeal microbiota. These preliminary and explorative results suggested that the neonatal microbial colonization is a highly dynamic process, which is sensitive to environmental factors.

However, several potential limitations should be taken into consideration. Firstly the sample amount might not be adequate. Secondly, if the characteristics of neonatal microbiota and maternal microbiota were studied together, it is more meaningful to know the initial source of microbial colonization.

In conclusion, the study's results suggest that, in some neonates, the microbiota may exist in both the intestine and the pharynx at birth. There was no significant difference between intestinal and pharyngeal microbiota composition on day 1, but on day 28, site-specific microbial was already established. Despite the initial similarity of pharyngeal and gut microbiotas, the specific bacterial genera were determined in both sites. Further we hypothesize that these genera may contribute to the functioning of the gut-lung axis.

Data Availability Statement

The original contributions presented in the study are included in the article/supplementary material, further inquiries can be directed to the corresponding author/s. https://figshare.com/s/81253de749d49bcb0898.

Ethics Statement

The studies involving human participants were reviewed and approved by Ethics Committee of West China Second University Hospital, Sichuan University. Written informed consent to participate in this study was provided by the participants' legal guardian/next of kin.

Author Contributions

SY: data collection, management and analysis, and manuscript drafting and revision. LQ and JS: data collection and manuscript revision. LX: site oversight and manuscript revision. YX and HL: study concept and design, interpretation of results, manuscript revision, and final approval. All authors contributed to the article and approved the submitted version.

Funding

This work was supported by the National Key R&D Program of China (2017YFC0211705), Key R&D Program of Science and Technology Department of Sichuan Province (19ZDYF1169), and Sichuan Science and Technology Program (2019YFH0143).

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

1. Munyaka PM, Khafipour E, Ghia JE. External influence of early childhood establishment of gut microbiota and subsequent health implications. Front Pediatr. (2014) 2:109. doi: 10.3389/fped.2014.00109

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Robertson RC, Manges AR, Finlay BB, Prendergast AJ. The human microbiome and child growth-first 1000 days and beyond. Trends Microbiol. (2019) 27:131–47. doi: 10.1016/j.tim.2018.09.008

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Li H, He Z, Gao D, Lv y, Zhou Q, Xiao B, et al. Characteristics of the intestinal microbiota in very low birth weight infants with extrauterine growth restriction. Front Pediatr. (2019) 7:99. doi: 10.3389/fped.2019.00099

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Masi AC, Stewart CJ. The role of the preterm intestinal microbiome in sepsis and necrotising enterocolitis. Early Hum Dev. (2019) 138:104854. doi: 10.1016/j.earlhumdev.2019.104854

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Subramanian S, Blanton LV, Frese SA, Charbonneau M, Mills DA, Gordon JI. Cultivating healthy growth and nutrition through the gut microbiota. Cell. (2015) 161:36–48. doi: 10.1016/j.cell.2015.03.013

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Zimmermann P, Messina N, Mohn WW, Finlay BB, Curtis N. Association between the intestinal microbiota and allergic sensitization, eczema, and asthma: a systematic review. J Allergy Clin Immunol. (2019) 143:467–85. doi: 10.1016/j.jaci.2018.09.025

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Qin J, Li Y, Cai Z, Li S, Zhu J, Zhang F, et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature. (2012) 490:55–60. doi: 10.1038/nature11450

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Li J, Zhao F, Wang Y, Chen J, Tao J, Tian G, et al. Gut microbiota dysbiosis contributes to the development of hypertension. Microbiome. (2017) 5:14. doi: 10.1186/s40168-016-0222-x

PubMed Abstract | CrossRef Full Text

9. Willyard C. Could baby's first bacteria take root before birth? Nature. (2018) 553:264–6. doi: 10.1038/d41586-018-00664-8

PubMed Abstract | CrossRef Full Text | Google Scholar

10. de Goffau MC, Lager S, Sovio U, Gaccioli F, Cook E, Peacock SJ, et al. Human placenta has no microbiome but can contain potential pathogens. Nature. (2019) 572:329–34. doi: 10.1038/s41586-019-1451-5

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Gentle SJ, Lal CV. Predicting BPD: lessons learned from the airway microbiome of preterm infants. Front Pediatr. (2020) 7:564. doi: 10.3389/fped.2019.00564

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Teo SM, Tang HHF, Mok D, Judd LM, Watts SC, Pham K, et al. Airway microbiota dynamics uncover a critical window for interplay of pathogenic bacteria and allergy in childhood respiratory disease. Cell Host Microbe. (2018) 24:341–52.e5. doi: 10.1016/j.chom.2018.08.005

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Chakradhar S. A curious connection: teasing apart the link between gut microbes and lung disease. Nat Med. (2017) 23:402–4. doi: 10.1038/nm0417-402

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Mjösberg J, Rao A. Lung inflammation originating in the gut. Science. (2018) 359:36–7. doi: 10.1126/science.aar4301

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Ranucci G, Buccigrossi V, de Freitas MB, Guarino A, Giannattasio A. Early-life intestine microbiota and lung health in children. J Immunol Res. (2017) 2017:8450496. doi: 10.1155/2017/8450496

PubMed Abstract | CrossRef Full Text | Google Scholar

16. He Y, Wen Q, Yao F, Xu D, Huang Y, Wang J. Gut-lung axis: the microbial contributions and clinical implications. Crit Rev Microbiol. (2017) 43:81–95. doi: 10.1080/1040841X.2016.1176988

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Surette MG, Bowdish DME. Loss of microbial topography precedes infection in infants. Am J Respir Crit Care Med. (2019) 200:660–2. doi: 10.1164/rccm.201903-0687ED

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Man WH, van Houten MA, Mérelle ME, Vlieger AM, Chu MLJN, Jansen NJG, et al. Bacterial and viral respiratory tract microbiota and host characteristics in children with lower respiratory tract infections: a matched case-control study. Lancet Respir Med. (2019) 7:417–26. doi: 10.1016/S2213-2600(18)30449-1

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Marsland BJ, Trompette A, Gollwitzer ES. The gut-lung axis in respiratory disease. Ann Am Thorac Soc. (2015) 12(Suppl. 2):S150–6.

Google Scholar

20. Chung KF. Airway microbial dysbiosis in asthmatic patients: a target for prevention and treatment? J Allergy Clin Immunol. (2017) 139:1071–81. doi: 10.1016/j.jaci.2017.02.004

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Qin N, Zheng B, Yao J, Guo L, Zuo J, Wu L, et al. Influence of H7N9 virus infection and associated treatment on human gut microbiota. Sci Rep. (2015) 5:14771. doi: 10.1038/srep14771

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Sokolowska M, Frei R, Lunjani N, Akdis CA, O'Mahony L. Microbiome and asthma. Asthma Res Pract. (2018) 4:1. doi: 10.1186/s40733-017-0037-y

CrossRef Full Text | Google Scholar

23. Hendrix MLE, van Kuijk SMJ, El Bahaey SE, Gerver WJM, Feron FJM, Kuin ME, et al. ‘Postnatal growth during the first five years of life in SGA and AGA neonates with reduced fetal growth'. Early Hum Dev. (2020) 151:105199. doi: 10.1016/j.earlhumdev.2020.105199

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Gurugubelli KR, Ballambattu VB, Bobby Z. Global DNA methylation in cord blood and neurodevelopmental outcome at 18 months of age among intrauterine growth restricted and appropriate for gestational age infants. J Trop Pediatr. (2020) fmaa108. doi: 10.1093/tropej/fmaa108

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Caporaso JG, Lauber CL, Walters WA, Berg-Lyons D, Lozupone CA, Turnbaugh PJ, et al. Global patterns of 16S rRNA diversity at a depth of millions of sequences per sample. Proc Natl Acad Sci USA. (2011) 108:4516–22. doi: 10.1073/pnas.1000080107

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Magoč T, Salzberg SL. FLASH: fast length adjustment of short reads to improve genome assemblies. Bioinformatics. (2011) 27:2957–63. doi: 10.1093/bioinformatics/btr507

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Wang Q, Garrity GM, Tiedje JM, Cole JR. Naive Bayesian classifier for rapid assignment of rRNA sequences into the new bacterial taxonomy. Appl Environ Microbiol. (2007) 73:5261–7. doi: 10.1128/AEM.00062-07

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Segata N, Izard J, Waldron L, Gevers D, Miropolsky L, Garrett WS, et al. Metagenomic biomarker discovery and explanation. Genome Biol. (2011) 12:R60. doi: 10.1186/gb-2011-12-6-r60

CrossRef Full Text

29. Fouhy F, Watkins C, Hill CJ, O'Shea CA, Nagle B, Dempsey EM, et al. Perinatal factors affect the gut microbiota up to four years after birth. Nat Commun. (2019) 10:1517. doi: 10.1038/s41467-019-09252-4

PubMed Abstract | CrossRef Full Text

30. Stockinger S, Hornef MW, Chassin C. Establishment of intestinal homeostasis during the neonatal period. Cell Mol Life Sci. (2011) 68:3699–712. doi: 10.1007/s00018-011-0831-2

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Jost T, Lacroix C, Braegger CP, Chassard C. New insights in gut microbiota establishment in healthy breast fed neonates. PLoS ONE. (2012) 7:e44595. doi: 10.1371/journal.pone.0044595

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Grier A, Qiu X, Bandyopadhyay S, Holden-Wiltse J, Kessler HA, Gill AL, et al. Impact of prematurity and nutrition on the developing gut microbiome and preterm infant growth. Microbiome. (2017) 5:158. doi: 10.1186/s40168-017-0377-0

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Faner R, Sibila O, Agustí A, Bernasconi E, Chalmers JD, Huffnagle GB, et al. The microbiome in respiratory medicine:current challenges and future perspectives. Eur Respir J. (2017) 49:1602086. doi: 10.1183/13993003.02086-2016

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Proctor DM, Relman DA. The landscape ecology and microbiota of the human nose, mouth, and throat. Cell Host Microbe. (2017) 21:421–32. doi: 10.1016/j.chom.2017.03.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: intestinal micriobiota, pharyngeal microbiota, 16S rRNA sequencing, preterm neonate, gut-lung axis

Citation: Yang S, Qiao L, Shi J, Xie L, Liu Y, Xiong Y and Liu H (2021) Clinical Study of Correlation for the Intestinal and Pharyngeal Microbiota in the Premature Neonates. Front. Pediatr. 9:632573. doi: 10.3389/fped.2021.632573

Received: 23 November 2020; Accepted: 18 January 2021;
Published: 16 February 2021.

Edited by:

Sinno Simons, Erasmus Medical Center, Netherlands

Reviewed by:

Grace Tzun-Wen Shaw, Academia Sinica, Taiwan
Valeriy Poroyko, Covance, United States
Ewa Skarżyńska, Medical University of Warsaw, Poland

Copyright © 2021 Yang, Qiao, Shi, Xie, Liu, Xiong and Liu. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Ying Xiong, xiongying246@163.com; Hanmin Liu, liuhm@scu.edu.cn

These authors have contributed equally to this work and share first authorship

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.