1
|
Xiang Q, Yan X, Shi X, Huang Y, Li L, Zhong J, Xu T, Tang S, Shi W, Zhou K. Prolonged premature rupture of membranes with increased risk of infection is associated with gut accumulation of Pseudomonas from the environment. Comput Struct Biotechnol J 2024; 23:2851-2860. [PMID: 39100803 PMCID: PMC11296040 DOI: 10.1016/j.csbj.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 07/04/2024] [Accepted: 07/05/2024] [Indexed: 08/06/2024] Open
Abstract
Background Preterm premature rupture of membranes (PPROM) contributes to over one-third of preterm births, and PPROM infants are more susceptible to infections. However, the risk factors remain poorly understood. We here aim to investigate the association of duration of premature rupture of membranes (PROM) and environmental microbiota with the gut microbiota and infection in PPROM infants. Methods Forty-six premature infants were recruited from two hospitals, and infant fecal and environmental samples were collected. 16 s rRNA sequencing was performed to analyze the fecal and environmental microbiome. Human inflammatory cytokines in cord vein plasma were measured. Results The gut microbiota composition of PPROM infants was different from that of non-PPROM infants, and the microbiome phenotypes were predicted to be associated with a higher risk of infection, further evidenced by the significantly increased levels of IL-6 and IL-8 in cord vein plasma of PPROM infants. The diversity of the gut microbiota in PPROM infants increased significantly as the duration of PROM excessed 12 h, and Pseudomonas contributed significantly to the dynamic changes. The Pseudomonas species in the gut of PPROM infants were highly homologous to those detected in the ward environment, suggesting that prolonged PROM is associated with horizontal transmission of environmental pathogens, leading to a higher risk of infection. Conclusions This study highlights that the duration of PROM is associated with the accumulation of environmental pathogens in the gut of PPROM infants, which is a risk factor for nosocomial infections. Improving environmental hygiene could be effective in optimizing the clinical care of PPROM infants.
Collapse
Affiliation(s)
- Quanhang Xiang
- Shenzhen Institute of Respiratory Diseases, the Second Clinical Medical College (Shenzhen People's Hospital), Jinan University ; The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China
| | - Xudong Yan
- Department of Neonatal Intensive Care Unit, the Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen 518020, China
| | - Xing Shi
- Shenzhen Institute of Respiratory Diseases, the Second Clinical Medical College (Shenzhen People's Hospital), Jinan University ; The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China
| | - Yi’e Huang
- Department of Prevention and Healthcare, Shenzhen Baoan Women’s and Children’s Hospital, Jinan University, Shenzhen 518020, China
| | - Lingfeng Li
- Shenzhen Institute of Respiratory Diseases, the Second Clinical Medical College (Shenzhen People's Hospital), Jinan University ; The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China
| | - Jiacheng Zhong
- Shenzhen Institute of Respiratory Diseases, the Second Clinical Medical College (Shenzhen People's Hospital), Jinan University ; The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China
| | - Tingting Xu
- Shenzhen Institute of Respiratory Diseases, the Second Clinical Medical College (Shenzhen People's Hospital), Jinan University ; The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China
| | - Shaohui Tang
- Department of Gastroenterology, the First Affiliated Hospital, Jinan University, Guangzhou 510632, China
| | - Wei Shi
- Department of Obstetrics, the Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen 518020, China
| | - Kai Zhou
- Shenzhen Institute of Respiratory Diseases, the Second Clinical Medical College (Shenzhen People's Hospital), Jinan University ; The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China
- Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen 518000, China
| |
Collapse
|
2
|
He J, Wang L, Ruan Y, Yan X, Liu Q, Chen B, Yang S, Du L. Comparison of intestinal and pharyngeal microbiota in preterm infants on the first day of life and the characteristics of pharyngeal microbiota in infants delivered by cesarean section or vaginally. Front Pediatr 2024; 12:1411887. [PMID: 39439450 PMCID: PMC11493734 DOI: 10.3389/fped.2024.1411887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 09/21/2024] [Indexed: 10/25/2024] Open
Abstract
Background This study aimed to explore the distribution of intestinal and pharyngeal microbiota on the first day of life in preterm infants and compare the composition of microbiota in infants delivered by cesarean section or vaginally. Methods This study included 44 late preterm infants with a gestational age of 34-36 + 6 weeks. Stool and throat swab samples were collected from the preterm infants on the first day of life. The infants were divided into cesarean section and vaginal delivery groups. Illumina NovaSeq high-throughput sequencing technology was used to sequence the V3-V4 hypervariable region of the 16S rRNA gene of all bacteria in the samples. Venn diagram was used to identify shared operational taxonomic units (OTUs) in the intestines and pharynges. Microbial analysis was conducted at the phylum and genus levels, and α and β diversity comparisons were performed. Results (1) Gestational age may have significantly affected the microbial colonization of the intestines and pharynges of preterm infants on the first day after birth (p ≤ 0.001). (2) More OTUs were detected in the pharynx than in the intestines, both have a total of 819 shared OTUs. Proteobacteria, Firmicutes, and Bacteroidota were the dominant phyla in both. At the genus level, Streptococcus had a lower relative abundance in stool samples (0.5%) compared to throat samples (0.5% vs. 22.2%, p = 0.003). 3) The relative abundance of Streptococcus in pharyngeal samples was 26.2% in the cesarean section group much higher than the 3.8% in the vaginal delivery group (p = 0.01). Conclusion The early postnatal period is a critical time for the establishment of an infant's microbiota. Gestational age at birth may influence microbial colonization, while birth weight, gender, and mode of delivery do not. The intestinal and pharyngeal microbiota composition of preterm infants on the first day after birth showed high similarity, but larger samples are needed for further validation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Sen Yang
- Department of Pediatrics, Chengdu Fifth People’s Hospital, Chengdu, China
| | - Lijun Du
- Department of Pediatrics, Chengdu Fifth People’s Hospital, Chengdu, China
| |
Collapse
|
3
|
Scheible K, Beblavy R, Sohn MB, Qui X, Gill AL, Narvaez-Miranda J, Brunner J, Miller RK, Barrett ES, O'Connor TG, Gill SR. Affective symptoms in pregnancy are associated with the vaginal microbiome. J Affect Disord 2024; 368:410-419. [PMID: 39293607 DOI: 10.1016/j.jad.2024.09.108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/11/2024] [Accepted: 09/14/2024] [Indexed: 09/20/2024]
Abstract
BACKGROUND Composition of the vaginal microbiome in pregnancy is associated with adverse maternal, obstetric, and child health outcomes. Therefore, identifying sources of individual differences in the vaginal microbiome is of considerable clinical and public health interest. The current study tested the hypothesis that vaginal microbiome composition during pregnancy is associated with an individual's experience of affective symptoms and stress exposure. METHODS Data were based on a prospective longitudinal study of a medically healthy community sample of 275 mother-infant pairs. Affective symptoms and stress exposure and select measures of associated biomarkers (diurnal salivary cortisol, serum measures of sex hormones) were collected at each trimester; self-report, clinical, and medical records were used to collect detailed data on socio-demographic factors and health behavior, including diet and sleep. Vaginal microbiome samples were collected in the third trimester (34-40 weeks) and characterized by 16S rRNA sequencing. Identified taxa were clustered into three community clusters (CC1-3) based on dissimilarity of vaginal microbiota composition. RESULTS Results indicate that depressive symptoms during pregnancy were reliably associated with individual taxa and CC3 in the third trimester. Prediction of functional potential from 16S taxonomy revealed a differential abundance of metabolic pathways in CC1-3 and individual taxa, including biosynthetic pathways for serotonin and dopamine. We did not find robust evidence linking symptom- and stress-related biomarkers and CCs. CONCLUSIONS Our results provide further evidence of how prenatal psychological distress during pregnancy alters the maternal-fetal microbiome ecosystem that may be important for understanding maternal and child health outcomes.
Collapse
Affiliation(s)
- Kristin Scheible
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA; Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Robert Beblavy
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Michael B Sohn
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Xing Qui
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Ann L Gill
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Janiret Narvaez-Miranda
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Jessica Brunner
- Department of Obstetrics and Gynecology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Richard K Miller
- Department of Obstetrics and Gynecology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Emily S Barrett
- Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ, USA; Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ, USA
| | - Thomas G O'Connor
- Department of Psychiatry, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA; Department of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA; Wynne Center for Family Research, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Steven R Gill
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| |
Collapse
|
4
|
Waller ME, Eichhorn CJ, Gutierrez A, Baatz JE, Wagner CL, Chetta KE, Engevik MA. Analyzing the Responses of Enteric Bacteria to Neonatal Intensive Care Supplements. Int J Microbiol 2024; 2024:3840327. [PMID: 39220439 PMCID: PMC11364479 DOI: 10.1155/2024/3840327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
In the neonatal intensive care unit, adequate nutrition requires various enteral products, including human milk and formula. Human milk is typically fortified to meet increased calorie goals, and infants commonly receive vitamin mixes, iron supplements, and less frequently, thickening agents. We examined the growth of 16 commensal microbes and 10 pathobionts found in the premature infant gut and found that formula, freshly pasteurized milk, and donated banked milk generally increased bacterial growth. Fortification of human milk significantly elevated the growth of all microbes. Supplementation with thickeners or NaCl in general did not stimulate additional growth. Vitamin mix promoted the growth of several commensals, while iron promoted growth of pathobionts. These data indicate that pathobionts in the preterm gut have significant growth advantage with preterm formula, fortified donor milk, and supplemented iron and suggest that the choice of milk and supplements may impact the infant gut microbiota.
Collapse
Affiliation(s)
- Megan E. Waller
- Department of Regenerative Medicine and Cell BiologyMedical University of South Carolina, Charleston, USA
| | - Caroline J. Eichhorn
- Department of Regenerative Medicine and Cell BiologyMedical University of South Carolina, Charleston, USA
| | - Alyssa Gutierrez
- Department of Regenerative Medicine and Cell BiologyMedical University of South Carolina, Charleston, USA
| | - John E. Baatz
- Department of PediatricsC.P. Darby Children's Research InstituteMedical University of South Carolina, Charleston, USA
- Department of PediatricsDivision of Neonatal-Perinatal MedicineMedical University of South CarolinaShawn Jenkins Children's Hospital, 10 McClennan Banks Drive, MSC 915, Charleston, SC 29425, USA
| | - Carol L. Wagner
- Department of PediatricsC.P. Darby Children's Research InstituteMedical University of South Carolina, Charleston, USA
- Department of PediatricsDivision of Neonatal-Perinatal MedicineMedical University of South CarolinaShawn Jenkins Children's Hospital, 10 McClennan Banks Drive, MSC 915, Charleston, SC 29425, USA
| | - Katherine E. Chetta
- Department of PediatricsC.P. Darby Children's Research InstituteMedical University of South Carolina, Charleston, USA
- Department of PediatricsDivision of Neonatal-Perinatal MedicineMedical University of South CarolinaShawn Jenkins Children's Hospital, 10 McClennan Banks Drive, MSC 915, Charleston, SC 29425, USA
| | - Melinda A. Engevik
- Department of Regenerative Medicine and Cell BiologyMedical University of South Carolina, Charleston, USA
- Department of Microbiology and ImmunologyMedical University of South Carolina, Charleston, USA
| |
Collapse
|
5
|
Jiang S, Cai M, Li D, Chen X, Chen X, Huang Q, Zhong C, Zheng X, Zhou D, Chen Z, Zhang L, Ching JY, Chen A, Lu S, Zhang L, Hu L, Liao Y, Li Y, He Z, Wu J, Huo H, Liang Y, Li W, Zou Y, Luo W, Ng SC, Chan FK, Chen X, Deng Y. Association of breast milk-derived arachidonic acid-induced infant gut dysbiosis with the onset of atopic dermatitis. Gut 2024:gutjnl-2024-332407. [PMID: 39084687 DOI: 10.1136/gutjnl-2024-332407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 06/27/2024] [Indexed: 08/02/2024]
Abstract
OBJECTIVE The specific breast milk-derived metabolites that mediate host-microbiota interactions and contribute to the onset of atopic dermatitis (AD) remain unknown and require further investigation. DESIGN We enrolled 250 mother-infant pairs and collected 978 longitudinal faecal samples from infants from birth to 6 months of age, along with 243 maternal faecal samples for metagenomics. Concurrently, 239 corresponding breast milk samples were analysed for metabolomics. Animal and cellular experiments were conducted to validate the bioinformatics findings. RESULTS The clinical findings suggested that a decrease in daily breastfeeding duration was associated with a reduced incidence of AD. This observation inspired us to investigate the effects of breast milk-derived fatty acids. We found that high concentrations of arachidonic acid (AA), but not eicosapentaenoic acid (EPA) or docosahexaenoic acid, induced gut dysbiosis in infants. Further investigation revealed that four specific bacteria degraded mannan into mannose, consequently enhancing the mannan-dependent biosynthesis of O-antigen and lipopolysaccharide. Correlation analysis confirmed that in infants with AD, the abundance of Escherichia coli under high AA concentrations was positively correlated with some microbial pathways (eg, 'GDP-mannose-derived O-antigen and lipopolysaccharide biosynthesis'). These findings are consistent with those of the animal studies. Additionally, AA, but not EPA, disrupted the ratio of CD4/CD8 cells, increased skin lesion area and enhanced the proportion of peripheral Th2 cells. It also promoted IgE secretion and the biosynthesis of prostaglandins and leukotrienes in BALB/c mice fed AA following ovalbumin immunostimulation. Moreover, AA significantly increased IL-4 secretion in HaCaT cells costimulated with TNF-α and INF-γ. CONCLUSIONS This study demonstrates that AA is intimately linked to the onset of AD via gut dysbiosis.
Collapse
Affiliation(s)
- Suhua Jiang
- Department of paediatrics, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Mengyun Cai
- Institute of translational medicine, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Dingru Li
- Institute of translational medicine, The First People's Hospital of Foshan, Foshan, People's Republic of China
- South China University of Technology School of Biology and Biological Engineering, Guangzhou, Guangdong, People's Republic of China
| | - Xiangping Chen
- Institute of translational medicine, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Xiaoqian Chen
- Department of paediatrics, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Qitao Huang
- Department of obstetrics, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Caimei Zhong
- Department of Dermatology, Shunde District Center for Prevention and Cure of Chronic Diseases, Foshan, China
| | - Xiufeng Zheng
- Department of Dermatology, Shunde Hospital, Southern Medical University, Lunjiao, Shunde, Foshan, People's Republic of China
| | - Dan Zhou
- Department of Breast Surgery, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Zhiyan Chen
- Department of Breast Surgery, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Lin Zhang
- Microbiota I-Center (MagIC), The Chinese University of Hong Kong, Hong Kong SAR, People's Republic of China
- Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR, People's Republic of China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, People's Republic of China
- Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinse University of Hong Kong, Hong Kong SAR, People's Republic of China
| | - Jessica Yl Ching
- Microbiota I-Center (MagIC), The Chinese University of Hong Kong, Hong Kong SAR, People's Republic of China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, People's Republic of China
| | - Ailing Chen
- Department of paediatrics, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Shaoxia Lu
- Department of obstetrics, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Lifang Zhang
- Institute of translational medicine, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Ling Hu
- Institute of translational medicine, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Yan Liao
- Department of obstetrics, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Ying Li
- Department of paediatrics, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Zhihua He
- Department of obstetrics, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Jingjing Wu
- Department of obstetrics, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Huiyi Huo
- Department of paediatrics, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Yongqi Liang
- Department of paediatrics, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Wanwen Li
- Department of paediatrics, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Yanli Zou
- The Second People's Hospital of Foshan, Foshan, People's Republic of China
| | - Wei Luo
- Institute of translational medicine, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Siew C Ng
- Microbiota I-Center (MagIC), The Chinese University of Hong Kong, Hong Kong SAR, People's Republic of China
- Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR, People's Republic of China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, People's Republic of China
- Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinse University of Hong Kong, Hong Kong SAR, People's Republic of China
| | - Francis Kl Chan
- Microbiota I-Center (MagIC), The Chinese University of Hong Kong, Hong Kong SAR, People's Republic of China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, People's Republic of China
| | - Xia Chen
- Central Laboratory of the Medical Research Center, The First Affiliated Hospital of Ningbo University, Ningbo, People's Republic of China
| | - Yuhua Deng
- Institute of translational medicine, The First People's Hospital of Foshan, Foshan, People's Republic of China
| |
Collapse
|
6
|
Wu X, Guo R, Fan Y, Chen S, Zheng W, Shu X, Chen B, Li X, Xu T, Shi L, Chen L, Shan L, Zhu Z, Tao E, Jiang M. Dynamic impact of delivery modes on gut microbiota in preterm infants hospitalized during the initial 4 weeks of life. Int J Med Microbiol 2024; 315:151621. [PMID: 38759506 DOI: 10.1016/j.ijmm.2024.151621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 03/05/2024] [Accepted: 05/05/2024] [Indexed: 05/19/2024] Open
Abstract
Preterm infants face a high risk of various complications, and their gut microbiota plays a pivotal role in health. Delivery modes have been reported to affect the development of gut microbiota in term infants, but its impact on preterm infants remains unclear. Here, we collected fecal samples from 30 preterm infants at five-time points within the first four weeks of life. Employing 16 S rRNA sequencing, principal coordinates analysis, the analysis of similarities, and the Wilcoxon rank-sum test, we examined the top dominant phyla and genera, the temporal changes in specific taxa abundance, and their relationship with delivery modes, such as Escherichia-Shigella and Enterococcus based on vaginal delivery and Pluralibacter related to cesarean section. Moreover, we identified particular bacteria, such as Taonella, Patulibacter, and others, whose proportions fluctuated among preterm infants born via different delivery modes at varying time points, as well as the microbiota types and functions. These results indicated the influence of delivery mode on the composition and function of the preterm infant gut microbiota. Importantly, these effects are time-dependent during the early stages of life. These insights shed light on the pivotal role of delivery mode in shaping the gut microbiota of preterm infants and have significant clinical implications for their care and management.
Collapse
Affiliation(s)
- Xin Wu
- Department of Pediatrics, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Rui Guo
- Gastrointestinal Laboratory and Pediatric Endoscopy Center, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, Zhejiang, China
| | - Yijia Fan
- Gastrointestinal Laboratory and Pediatric Endoscopy Center, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, Zhejiang, China
| | - Shuang Chen
- Gastrointestinal Laboratory and Pediatric Endoscopy Center, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, Zhejiang, China
| | - Wei Zheng
- Gastrointestinal Laboratory and Pediatric Endoscopy Center, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, Zhejiang, China
| | - Xiaoli Shu
- Gastrointestinal Laboratory and Pediatric Endoscopy Center, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, Zhejiang, China
| | - Bo Chen
- Gastrointestinal Laboratory and Pediatric Endoscopy Center, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, Zhejiang, China
| | - Xing Li
- Department of Pediatrics, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Tingting Xu
- Department of Pediatrics, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Lingbing Shi
- Department of Pediatrics, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Li Chen
- Department of Pediatrics, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Lichun Shan
- Department of Pediatrics, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Zhenya Zhu
- Gastrointestinal Laboratory and Pediatric Endoscopy Center, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, Zhejiang, China
| | - Enfu Tao
- Department of Neonatology and NICU, Wenling Maternal and Child Health Care Hospital, Wenling, Zhejiang Province, China
| | - Mizu Jiang
- Gastrointestinal Laboratory and Pediatric Endoscopy Center, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, Zhejiang, China; Department of Gastroenterology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, Zhejiang, China.
| |
Collapse
|
7
|
Dombrowska-Pali A, Wiktorczyk-Kapischke N, Chrustek A, Olszewska-Słonina D, Gospodarek-Komkowska E, Socha MW. Human Milk Microbiome-A Review of Scientific Reports. Nutrients 2024; 16:1420. [PMID: 38794658 PMCID: PMC11124344 DOI: 10.3390/nu16101420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/01/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
One of the most important bioactive components of breast milk are free breast milk oligosaccharides, which are a source of energy for commensal intestinal microorganisms, stimulating the growth of Bifidobacterium, Lactobacillus, and Bacteroides in a child's digestive tract. There is some evidence that maternal, perinatal, and environmental-cultural factors influence the modulation of the breast milk microbiome. This review summarizes research that has examined the composition of the breast milk microbiome and the factors that may influence it. The manuscript highlights the potential importance of the breast milk microbiome for the future development and health of children. The origin of bacteria in breast milk is thought to include the mother's digestive tract (entero-mammary tract), bacterial exposure to the breast during breastfeeding, and the retrograde flow of breast milk from the infant's mouth to the woman's milk ducts. Unfortunately, despite increasingly more precise methods for assessing microorganisms in human milk, the topic of the human milk microbiome is still quite limited and requires scientific research that takes into account various conditions.
Collapse
Affiliation(s)
- Agnieszka Dombrowska-Pali
- Department of Perinatology, Gynecology and Gynecologic Oncology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Łukasiewicza 1, 85-821 Bydgoszcz, Poland;
| | - Natalia Wiktorczyk-Kapischke
- Department of Microbiology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie-Skłodowskiej 9, 85-094 Bydgoszcz, Poland; (N.W.-K.); (E.G.-K.)
| | - Agnieszka Chrustek
- Department of Pathobiochemistry and Clinical Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie-Skłodowskiej 9, 85-094 Bydgoszcz, Poland; (A.C.); (D.O.-S.)
| | - Dorota Olszewska-Słonina
- Department of Pathobiochemistry and Clinical Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie-Skłodowskiej 9, 85-094 Bydgoszcz, Poland; (A.C.); (D.O.-S.)
| | - Eugenia Gospodarek-Komkowska
- Department of Microbiology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie-Skłodowskiej 9, 85-094 Bydgoszcz, Poland; (N.W.-K.); (E.G.-K.)
| | - Maciej W. Socha
- Department of Perinatology, Gynecology and Gynecologic Oncology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Łukasiewicza 1, 85-821 Bydgoszcz, Poland;
- Department of Obstetrics and Gynecology, St. Adalberts’s Hospital in Gdańsk, Copernicus Healthcare Entity LLC, Jana Pawła II 50, 80-462 Gdańsk, Poland
| |
Collapse
|
8
|
Younge N. Influence of infant microbiome on health and development. Clin Exp Pediatr 2024; 67:224-231. [PMID: 37605538 PMCID: PMC11065641 DOI: 10.3345/cep.2023.00598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/08/2023] [Accepted: 07/19/2023] [Indexed: 08/23/2023] Open
Abstract
The microbiome is a complex ecosystem comprising microbes, their genomes, and the surrounding environment. The microbiome plays a critical role in early human development, including maturation of the host immune system and gastrointestinal tract. Multiple factors, including diet, anti-biotic use, and other environmental exposures, influence the establishment of the microbiome during infancy. Numerous studies have identified associations between the microbiome and neonatal diseases, including necrotizing enterocolitis, sepsis, and malnutrition. Furthermore, there is compelling evidence that perturbation of the microbiome in early life can have lasting developmental effects that increase an individual's risk for immune and metabolic diseases in later life. Supplementation of the microbiome with probiotics reduces the risk of necrotizing enterocolitis and sepsis in at-risk infants. This review focuses on the structure and function of the infant microbiome, the environmental and clinical factors that influence its assembly, and its impact on infant health and development.
Collapse
|
9
|
Daniels BN, Nurge J, De Smet C, Sleeper O, White C, Davidson JM, Fidopiastis P. Microbiome composition and function within the Kellet's whelk perivitelline fluid. Microbiol Spectr 2024; 12:e0351423. [PMID: 38334378 PMCID: PMC10913743 DOI: 10.1128/spectrum.03514-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 01/17/2024] [Indexed: 02/10/2024] Open
Abstract
Microbiomes have gained significant attention in ecological research, owing to their diverse interactions and essential roles within different organismal ecosystems. Microorganisms, such as bacteria, archaea, and viruses, have profound impact on host health, influencing digestion, metabolism, immune function, tissue development, and behavior. This study investigates the microbiome diversity and function of Kellet's whelk (Kelletia kelletii) perivitelline fluid (PVF), which sustains thousands of developing K. kelletii embryos within a polysaccharide and protein matrix. Our core microbiome analysis reveals a diverse range of bacteria, with the Roseobacter genus being the most abundant. Additionally, genes related to host-microbe interactions, symbiosis, and quorum sensing were detected, indicating a potential symbiotic relationship between the microbiome and Kellet's whelk embryos. Furthermore, the microbiome exhibits gene expression related to antibiotic biosynthesis, suggesting a defensive role against pathogenic bacteria and potential discovery of novel antibiotics. Overall, this study sheds light on the microbiome's role in Kellet's whelk development, emphasizing the significance of host-microbe interactions in vulnerable life history stages. To our knowledge, ours is the first study to use 16S sequencing coupled with RNA sequencing (RNA-seq) to profile the microbiome of an invertebrate PVF.IMPORTANCEThis study provides novel insight to an encapsulated system with strong evidence of symbiosis between the microbial inhabitants and developing host embryos. The Kellet's whelk perivitelline fluid (PVF) contains microbial organisms of interest that may be providing symbiotic functions and potential antimicrobial properties during this vulnerable life history stage. This study, the first to utilize a comprehensive approach to investigating Kellet's whelk PVF microbiome, couples 16S rRNA gene long-read sequencing with RNA-seq. This research contributes to and expands our knowledge on the roles of beneficial host-associated microbes.
Collapse
Affiliation(s)
- Benjamin N. Daniels
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, California, USA
| | - Jenna Nurge
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, California, USA
| | - Chanel De Smet
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, California, USA
| | - Olivia Sleeper
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, California, USA
| | - Crow White
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, California, USA
| | - Jean M. Davidson
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, California, USA
| | - Pat Fidopiastis
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, California, USA
| |
Collapse
|
10
|
David P, Claud EC. Necrotizing Enterocolitis and the Preterm Infant Microbiome. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1449:29-41. [PMID: 39060729 DOI: 10.1007/978-3-031-58572-2_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
Preterm infants differ significantly from their term infant counterparts regarding bacterial colonization patterns related to maternal microbiota diversity, mode of delivery, feeding type, antibiotic exposure, and the environmental influences related to prolonged hospitalization in the neonatal intensive care unit (NICU). Necrotizing enterocolitis (NEC), a multifactorial intestinal disorder characterized by ischemic bowel disease, disproportionately impacts preterm infants and has a high disease burden. Recent studies in the basic, translational, and clinical scientific literature have advanced knowledge into this complex disease process. Despite the explosion of research into NEC, however, there is a still a great deal unknown about this devastating illness. Additionally, the disease morbidity and mortality for NEC remain high despite advances in therapy options. This chapter reviews the current literature into the preterm infant microbiome, pathogenesis of NEC, potential targets for altering preterm microbiome, influence of microbiome on other organ systems, long-term implications of microbiome dysbiosis, and future directions of study.
Collapse
Affiliation(s)
- Pyone David
- Department of Pediatrics, Pritzker School of Medicine, The University of Chicago, Chicago, IL, USA.
| | - Erika C Claud
- Department of Pediatrics, Pritzker School of Medicine, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
11
|
DuPont HL, Salge MMH. The Importance of a Healthy Microbiome in Pregnancy and Infancy and Microbiota Treatment to Reverse Dysbiosis for Improved Health. Antibiotics (Basel) 2023; 12:1617. [PMID: 37998819 PMCID: PMC10668833 DOI: 10.3390/antibiotics12111617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 11/05/2023] [Accepted: 11/09/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND The microbiome of newborn infants during the first 1000 days, influenced early on by their mothers' microbiome health, mode of delivery and breast feeding, orchestrates the education and programming of the infant's immune system and determines in large part the general health of the infant for years. METHODS PubMed was reviewed for maternal infant microbiome health and microbiota therapy in this setting with prebiotics, probiotics, vaginal seeding and fecal microbiota transplantation (FMT). RESULTS A healthy nonobese mother, vaginal delivery and strict breast feeding contribute to microbiome health in a newborn and young infant. With reduced microbiome diversity (dysbiosis) during pregnancy, cesarean delivery, prematurity, and formula feeding contribute to dysbiosis in the newborn. Microbiota therapy is an important approach to repair dysbiosis in pregnant women and their infants. Currently available probiotics can have favorable metabolic effects on mothers and infants, but these effects are variable. In research settings, reversal of infant dysbiosis can be achieved via vaginal seeding or FMT. Next generation probiotics in development should replace current probiotics and FMT. CONCLUSIONS The most critical phase of human microbiome development is in the first 2-3 years of life. Preventing and treating dysbiosis during pregnancy and early life can have a profound effect on an infant's later health.
Collapse
Affiliation(s)
- Herbert L. DuPont
- Division of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas, Houston, TX 77030, USA
- Department of Internal Medicine, University of Texas McGovern Medical School, Houston, TX 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Kelsey Research Foundation, Houston, TX 77005, USA
| | | |
Collapse
|
12
|
Yu J, Jiang C, Yamano R, Koike S, Sakai Y, Mino S, Sawabe T. Unveiling the early life core microbiome of the sea cucumber Apostichopus japonicus and the unexpected abundance of the growth-promoting Sulfitobacter. Anim Microbiome 2023; 5:54. [PMID: 37876012 PMCID: PMC10599069 DOI: 10.1186/s42523-023-00276-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 10/16/2023] [Indexed: 10/26/2023] Open
Abstract
BACKGROUND Microbiome in early life has long-term effects on the host's immunological and physiological development and its disturbance is known to trigger various diseases in host Deuterostome animals. The sea cucumber Apostichopus japonicus is one of the most valuable marine Deuterostome invertebrates in Asia and a model animal in regeneration studies. To understand factors that impact on host development and holobiont maintenance, host-microbiome association has been actively studied in the last decade. However, we currently lack knowledge of early life core microbiome during its ontogenesis and how it benefits the host's growth. RESULTS We analyzed the microbial community in 28 sea cucumber samples from a laboratory breeding system, designed to replicate aquaculture environments, across six developmental stages (fertilized eggs to the juvenile stage) over a three years-period to examine the microbiomes' dynamics and stability. Microbiome shifts occurred during sea cucumber larval ontogenesis in every case. Application of the most sophisticated core microbiome extraction methodology, a hybrid approach with abundance-occupancy core microbiome analyses (top 75% of total reads and > 70% occupation) and core index calculation, first revealed early life core microbiome consisted of Alteromonadaceae and Rhodobacteraceae, as well as a stage core microbiome consisting of pioneer core microbe Pseudoalteromonadaceae in A. japonicus, suggesting a stepwise establishment of microbiome related to ontogenesis and feeding behavior in A. japonicus. More interestingly, four ASVs affiliated to Alteromonadaceae and Rhodobacteraceae were extracted as early life core microbiome. One of the ASV (ASV0007) was affiliated to the Sulfitobactor strain BL28 (Rhodobacteraceae), isolated from blastula larvae in the 2019 raring batch. Unexpectedly, a bioassay revealed the BL28 strain retains a host growth-promoting ability. Further meta-pangenomics approach revealed the BL28 genome reads were abundant in the metagenomic sequence pool, in particular, in that of post-gut development in early life stages of A. japonicus. CONCLUSION Repeated rearing efforts of A. japonicus using laboratory aquaculture replicating aquaculture environments and hybrid core microbiome extraction approach first revealed particular ASVs affiliated to Alteromonadaceae and Rhodobacteraceae as the A. japonicus early life core microbiome. Further bioassay revealed the growth promoting ability to the host sea cucumber in one of the core microbes, the Sulfitobactor strain BL28 identified as ASV0007. Genome reads of the BL28 were abundant in post-gut development of A. japonicus, which makes us consider effective probiotic uses of those core microbiome for sea cucumber resource production and conservation. The study also emphasizes the importance of the core microbiome in influencing early life stages in marine invertebrates. Understanding these dynamics could offer pathways to improve growth, immunity, and disease resistance in marine invertebrates.
Collapse
Affiliation(s)
- Juanwen Yu
- Laboratory of Microbiology, Faculty of Fisheries Sciences, Hokkaido University, Hakodate, Japan.
| | - Chunqi Jiang
- Laboratory of Microbiology, Faculty of Fisheries Sciences, Hokkaido University, Hakodate, Japan
- Atmosphere and Ocean Research Institute, The University of Tokyo, Chiba, Japan
| | - Ryota Yamano
- Laboratory of Microbiology, Faculty of Fisheries Sciences, Hokkaido University, Hakodate, Japan
| | - Shotaro Koike
- Laboratory of Microbiology, Faculty of Fisheries Sciences, Hokkaido University, Hakodate, Japan
| | - Yuichi Sakai
- Hakodate Fisheries Research, Hokkaido Research Organization, Local Independent Administrative Agency, Hakodate, Japan
| | - Sayaka Mino
- Laboratory of Microbiology, Faculty of Fisheries Sciences, Hokkaido University, Hakodate, Japan
| | - Tomoo Sawabe
- Laboratory of Microbiology, Faculty of Fisheries Sciences, Hokkaido University, Hakodate, Japan.
| |
Collapse
|
13
|
Beharry KD, Latkowska M, Valencia AM, Allana A, Soto J, Cai CL, Golombek S, Hand I, Aranda JV. Factors Influencing Neonatal Gut Microbiome and Health with a Focus on Necrotizing Enterocolitis. Microorganisms 2023; 11:2528. [PMID: 37894186 PMCID: PMC10608807 DOI: 10.3390/microorganisms11102528] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/21/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Maturational changes in the gut start in utero and rapidly progress after birth, with some functions becoming fully developed several months or years post birth including the acquisition of a full gut microbiome, which is made up of trillions of bacteria of thousands of species. Many factors influence the normal development of the neonatal and infantile microbiome, resulting in dysbiosis, which is associated with various interventions used for neonatal morbidities and survival. Extremely low gestational age neonates (<28 weeks' gestation) frequently experience recurring arterial oxygen desaturations, or apneas, during the first few weeks of life. Apnea, or the cessation of breathing lasting 15-20 s or more, occurs due to immature respiratory control and is commonly associated with intermittent hypoxia (IH). Chronic IH induces oxygen radical diseases of the neonate, including necrotizing enterocolitis (NEC), the most common and devastating gastrointestinal disease in preterm infants. NEC is associated with an immature intestinal structure and function and involves dysbiosis of the gut microbiome, inflammation, and necrosis of the intestinal mucosal layer. This review describes the factors that influence the neonatal gut microbiome and dysbiosis, which predispose preterm infants to NEC. Current and future management and therapies, including the avoidance of dysbiosis, the use of a human milk diet, probiotics, prebiotics, synbiotics, restricted antibiotics, and fecal transplantation, for the prevention of NEC and the promotion of a healthy gut microbiome are also reviewed. Interventions directed at boosting endogenous and/or exogenous antioxidant supplementation may not only help with prevention, but may also lessen the severity or shorten the course of the disease.
Collapse
Affiliation(s)
- Kay D. Beharry
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA; (M.L.); (C.L.C.); (S.G.); (J.V.A.)
| | - Magdalena Latkowska
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA; (M.L.); (C.L.C.); (S.G.); (J.V.A.)
| | - Arwin M. Valencia
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Saddleback Memorial Medical Center, Laguna Hills, CA 92653, USA;
| | - Ahreen Allana
- Department of Pediatrics, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA; (A.A.); (J.S.)
| | - Jatnna Soto
- Department of Pediatrics, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA; (A.A.); (J.S.)
| | - Charles L. Cai
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA; (M.L.); (C.L.C.); (S.G.); (J.V.A.)
| | - Sergio Golombek
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA; (M.L.); (C.L.C.); (S.G.); (J.V.A.)
| | - Ivan Hand
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Kings County Hospital Center, Brooklyn, NY 11203, USA;
| | - Jacob V. Aranda
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA; (M.L.); (C.L.C.); (S.G.); (J.V.A.)
| |
Collapse
|
14
|
Xiang Q, Yan X, Shi W, Li H, Zhou K. Early gut microbiota intervention in premature infants: Application perspectives. J Adv Res 2023; 51:59-72. [PMID: 36372205 PMCID: PMC10491976 DOI: 10.1016/j.jare.2022.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/30/2022] [Accepted: 11/05/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Preterm birth is the leading cause of death in children under the age of five. One of the major factors contributing to the high risk of diseases and deaths in premature infants is the incomplete development of the intestinal immune system. The gut microbiota has been widely recognized as a critical factor in promoting the development and function of the intestinal immune system after birth. However, the gut microbiota of premature infants is at high risk of dysbiosis, which is highly associated with adverse effects on the development and education of the early life immune system. Early intervention can modulate the colonization and development of gut microbiota and has a long-term influence on the development of the intestinal immune system. AIM OF REVIEW This review aims to summarize the characterization, interconnection, and underlying mechanism of gut microbiota and intestinal innate immunity in premature infants, and to discuss the status, applicability, safety, and prospects of different intervention strategies in premature infants, thus providing an overview and outlook of the current applications and remaining gaps of early intervention strategies in premature infants. KEY SCIENTIFIC CONCEPTS OF REVIEW This review is focused on three key concepts. Firstly, the gut microbiota of premature infants is at high risk of dysbiosis, resulting in dysfunctional intestinal immune system processes. Secondly, contributing roles of early intervention have been observed in improving the intestinal environment and promoting gut microbiota colonization, which is significant in the development and function of gut immunity in premature infants. Thirdly, different strategies of early intervention, such as probiotics, fecal microbiota transplantation, and nutrients, show different safety, applicability, and outcome in premature infants, and the underlying mechanism is complex and poorly understood.
Collapse
Affiliation(s)
- Quanhang Xiang
- Shenzhen Institute of Respiratory Diseases, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Xudong Yan
- Department of Neonatal Intensive Care Unit, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Wei Shi
- Department of Obstetrics and Gynecology, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Huiping Li
- Department of Respiratory and Critical Care Medicine, the first affiliated hospital of Southern University of Science and Technology of China, Shenzhen People's Hospital, Shenzhen, China; The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China
| | - Kai Zhou
- Shenzhen Institute of Respiratory Diseases, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China; The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
15
|
Letourneau J, Walker L, Han SH, David LA, Younge N. Fecal pH and redox as functional markers in the premature infant gut microbiome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.14.553216. [PMID: 37645803 PMCID: PMC10462032 DOI: 10.1101/2023.08.14.553216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
The infant gut microbiome is a crucial factor in health and development. In preterm infants, altered gut microbiome composition and function have been linked to serious neonatal complications such as necrotizing enterocolitis and sepsis, which can lead to long-term disability. Although many studies have described links between microbiome composition and disease risk, there is a need for biomarkers to identify infants at risk of these complications in practice. In this study, we obtained stool samples from preterm infant participants longitudinally during the first postnatal months, and measured pH and redox, as well as SCFA content and microbiome composition by 16S rRNA gene amplicon sequencing. These outcomes were compared to clinical data to better understand the role of pH and redox in infant gut microbiome development and overall health, and to assess the potential utility of pH and redox as biomarkers. We found that infants born earlier or exposed to antibiotics exhibited increased fecal pH, and that redox potential increased with postnatal age. These differences may be linked to changes in SCFA content, which was correlated with pH and increased with age. Microbiome composition was also related to birth weight, age, pH, and redox. Our findings suggest that pH and redox may serve as biomarkers of metabolic state in the preterm infant gut.
Collapse
Affiliation(s)
- Jeffrey Letourneau
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27708
| | - LaShawndra Walker
- Department of Pediatrics, Duke University School of Medicine, Durham, NC 27708
| | - Se Hyang Han
- Department of Pediatrics, Duke University School of Medicine, Durham, NC 27708
| | - Lawrence A David
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27708
- Program in Computational Biology and Bioinformatics, Duke University School of Medicine, Durham, NC 27708
| | - Noelle Younge
- Department of Pediatrics, Duke University School of Medicine, Durham, NC 27708
| |
Collapse
|
16
|
Kartjito MS, Yosia M, Wasito E, Soloan G, Agussalim AF, Basrowi RW. Defining the Relationship of Gut Microbiota, Immunity, and Cognition in Early Life-A Narrative Review. Nutrients 2023; 15:2642. [PMID: 37375546 DOI: 10.3390/nu15122642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/22/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Recently, the immune system has been identified as one of the possible main bridges which connect the gut-brain axis. This review aims to examine available evidence on the microbiota-immunity-cognitive relationship and its possible effects on human health early in life. This review was assembled by compiling and analyzing various literature and publications that document the gut microbiota-immune system-cognition interaction and its implications in the pediatric population. This review shows that the gut microbiota is a pivotal component of gut physiology, with its development being influenced by a variety of factors and, in return, supports the development of overall health. Findings from current research focus on the complex relationship between the central nervous system, gut (along with gut microbiota), and immune cells, highlighting the importance of maintaining a balanced interaction among these systems for preserving homeostasis, and demonstrating the influence of gut microbes on neurogenesis, myelin formation, the potential for dysbiosis, and alterations in immune and cognitive functions. While limited, evidence shows how gut microbiota affects innate and adaptive immunity as well as cognition (through HPA axis, metabolites, vagal nerve, neurotransmitter, and myelination).
Collapse
Affiliation(s)
| | - Mikhael Yosia
- Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
| | - Erika Wasito
- Medical and Science Affairs Division, Danone Specialized Nutrition Indonesia, Jakarta 12950, Indonesia
| | - Garry Soloan
- Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
| | | | - Ray Wagiu Basrowi
- Medical and Science Affairs Division, Danone Specialized Nutrition Indonesia, Jakarta 12950, Indonesia
| |
Collapse
|
17
|
Pace E, Yanowitz TD, Waltz P, Morowitz MJ. Antibiotic therapy and necrotizing enterocolitis. Semin Pediatr Surg 2023; 32:151308. [PMID: 37295297 DOI: 10.1016/j.sempedsurg.2023.151308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Antibiotic therapy remains a cornerstone of treatment of both medical and surgical presentations of necrotizing enterocolitis (NEC). However, guidelines regarding the administration of antibiotics for the treatment of NEC are lacking and practices vary amongst clinicians. Although the pathogenesis of NEC is unknown, there is consensus that the infant gastrointestinal microbiome contributes to the disease. The presumed connection between dysbiosis and NEC has prompted some to study whether early prophylactic enteral antibiotics can prevent NEC. Yet others have taken an opposing approach, studying whether perinatal antibiotic exposure increases the risk of NEC by inducing a state of dysbiosis. This narrative review summarizes what is known about antibiotics and their association with the infant microbiome and NEC, current antibiotic prescribing practices for infants with medical and surgical NEC, as well as potential strategies to further optimize the use of antibiotics in this population of infants.
Collapse
Affiliation(s)
- Elizabeth Pace
- University of Pittsburgh Department of Surgery, United States
| | - Toby D Yanowitz
- University of Pittsburgh Department of Pediatrics, Division of Neonatology, United States
| | - Paul Waltz
- University of Pittsburgh Department of Surgery, Division of Pediatric General and Thoracic Surgery, UPMC Children's Hospital of Pittsburgh, 4401 Penn Ave., Pittsburgh, PA 15224, United States
| | - Michael J Morowitz
- University of Pittsburgh Department of Surgery, Division of Pediatric General and Thoracic Surgery, UPMC Children's Hospital of Pittsburgh, 4401 Penn Ave., Pittsburgh, PA 15224, United States.
| |
Collapse
|
18
|
Wilson A, Bogie B, Chaaban H, Burge K. The Nonbacterial Microbiome: Fungal and Viral Contributions to the Preterm Infant Gut in Health and Disease. Microorganisms 2023; 11:909. [PMID: 37110332 PMCID: PMC10144239 DOI: 10.3390/microorganisms11040909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/27/2023] [Accepted: 03/30/2023] [Indexed: 04/29/2023] Open
Abstract
The intestinal microbiome is frequently implicated in necrotizing enterocolitis (NEC) pathogenesis. While no particular organism has been associated with NEC development, a general reduction in bacterial diversity and increase in pathobiont abundance has been noted preceding disease onset. However, nearly all evaluations of the preterm infant microbiome focus exclusively on the bacterial constituents, completely ignoring any fungi, protozoa, archaea, and viruses present. The abundance, diversity, and function of these nonbacterial microbes within the preterm intestinal ecosystem are largely unknown. Here, we review findings on the role of fungi and viruses, including bacteriophages, in preterm intestinal development and neonatal intestinal inflammation, with potential roles in NEC pathogenesis yet to be determined. In addition, we highlight the importance of host and environmental influences, interkingdom interactions, and the role of human milk in shaping fungal and viral abundance, diversity, and function within the preterm intestinal ecosystem.
Collapse
Affiliation(s)
| | | | - Hala Chaaban
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Kathryn Burge
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
19
|
Neves LL, Hair AB, Preidis GA. A systematic review of associations between gut microbiota composition and growth failure in preterm neonates. Gut Microbes 2023; 15:2190301. [PMID: 36927287 PMCID: PMC10026866 DOI: 10.1080/19490976.2023.2190301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
Growth failure is among the most prevalent and devastating consequences of prematurity. Up to half of all extremely preterm neonates struggle to grow despite modern nutrition practices. Although elegant preclinical models suggest causal roles for the gut microbiome, these insights have not yet translated into biomarkers that identify at-risk neonates or therapies that prevent or treat growth failure. This systematic review aims to identify features of the neonatal gut microbiota that are positively or negatively associated with early postnatal growth. We identified 860 articles, of which 14 were eligible for inclusion. No two studies used the same definitions of growth, ages at stool collection, and statistical methods linking microbiota to metadata. In all, 58 different taxa were associated with growth, with little consensus among studies. Two or more studies reported positive associations with Enterobacteriaceae, Bacteroides, Bifidobacterium, Enterococcus, and Veillonella, and negative associations with Citrobacter, Klebsiella, and Staphylococcus. Streptococcus was positively associated with growth in five studies and negatively associated with growth in three studies. To gain insight into how the various definitions of growth could impact results, we performed an exploratory secondary analysis of 245 longitudinally sampled preterm infant stools, linking microbiota composition to multiple clinically relevant definitions of neonatal growth. Within this cohort, every definition of growth was associated with a different combination of microbiota features. Together, these results suggest that the lack of consensus in defining neonatal growth may limit our capacity to detect consistent, meaningful clinical associations that could be leveraged into improved care for preterm neonates.
Collapse
Affiliation(s)
- Larissa L. Neves
- Division of Gastroenterology, Hepatology, & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| | - Amy B. Hair
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| | - Geoffrey A. Preidis
- Division of Gastroenterology, Hepatology, & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| |
Collapse
|
20
|
Wang X, Geng S. Diet-gut microbial interactions influence cancer immunotherapy. Front Oncol 2023; 13:1138362. [PMID: 37035188 PMCID: PMC10081683 DOI: 10.3389/fonc.2023.1138362] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 03/16/2023] [Indexed: 04/11/2023] Open
Abstract
The gut microbiome is involved in the absorption and metabolism of host nutrients and modulates the immune response, affecting the efficacy of immunotherapy for cancer. In patients receiving immunotherapy, appropriate modifications of gut microbiota are thought to improve therapeutic response. Of all the factors that influence the gut microbiota, diet is the most influential and modifiable. Healthy dietary patterns as well as some specific dietary components can help the growth of beneficial microbiota in the gut, thereby protecting against cancers and promoting human health. A growing number of researches have confirmed the positive effects of a diet-gut microbiota approach as an adjuvant therapy for cancer, but controversy remains. Here, we summarize the interactions between diet and gut microbes based on previous studies, and discuss the role of gut microbiota-based dietary strategies in tumor immunotherapy, with the potential mechanisms of actions also intensively discussed.
Collapse
Affiliation(s)
- Xue Wang
- Department of Oncology, First People's Hospital of Guangyuan, Guangyuan, China
| | - Shitao Geng
- Department of Emergency, First Naval Hospital of Southern Theater Command, Zhanjiang, China
| |
Collapse
|
21
|
Lynch LE, Hair AB, Soni KG, Yang H, Gollins LA, Narvaez-Rivas M, Setchell KDR, Preidis GA. Cholestasis impairs gut microbiota development and bile salt hydrolase activity in preterm neonates. Gut Microbes 2023; 15:2183690. [PMID: 36843227 PMCID: PMC9980517 DOI: 10.1080/19490976.2023.2183690] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/28/2023] Open
Abstract
Cholestasis refers to impaired bile flow from the liver to the intestine. In neonates, cholestasis causes poor growth and may progress to liver failure and death. Normal bile flow requires an intact liver-gut-microbiome axis, whereby liver-derived primary bile acids are transformed into secondary bile acids. Microbial bile salt hydrolase (BSH) enzymes are responsible for the first step, deconjugating glycine- and taurine-conjugated primary bile acids. Cholestatic neonates often are treated with the potent choleretic bile acid ursodeoxycholic acid (UDCA), although interactions between UDCA, gut microbes, and other bile acids are poorly understood. To gain insight into how the liver-gut-microbiome axis develops in extreme prematurity and how cholestasis alters this maturation, we conducted a nested case-control study collecting 124 stool samples longitudinally from 24 preterm infants born at mean 27.2 ± 1.8 weeks gestation and 946 ± 249.6 g, half of whom developed physiologic cholestasis. Samples were analyzed by whole metagenomic sequencing, in vitro BSH enzyme activity assays optimized for low biomass fecal samples, and quantitative mass spectrometry to measure the bile acid metabolome. In extremely preterm neonates, acquisition of the secondary bile acid biosynthesis pathway and BSH genes carried by Clostridium perfringens are the most prominent features of early microbiome development. Cholestasis interrupts this developmental pattern. BSH gene abundance and enzyme activity are profoundly reduced in cholestatic neonates, resulting in decreased quantities of unconjugated bile acids. UDCA restores total fecal bile acid levels in cholestatic neonates, but this is due to a 522-fold increase in fecal UDCA. A majority of bile acids in early development are atypical positional and stereo-isomers of bile acids. We report novel associations linking isomeric bile acids and BSH activity to neonatal growth trajectories. These data highlight deconjugation of bile acids as a key microbial function that is acquired in early neonatal development and impaired by cholestasis.
Collapse
Affiliation(s)
- Lauren E. Lynch
- Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| | - Amy B. Hair
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA,CONTACT Amy B. Hair Division of Neonatology, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, 6621 Fannin Street, Suite A5590, Houston, TX77030, USA
| | - Krishnakant G. Soni
- Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| | - Heeju Yang
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| | - Laura A. Gollins
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| | - Monica Narvaez-Rivas
- Division of Pathology and Laboratory Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Kenneth D. R. Setchell
- Division of Pathology and Laboratory Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Geoffrey A. Preidis
- Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA,Geoffrey A. Preidis Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, 1102 Bates Avenue, Feigin Tower Suite 860, Houston, TX77030, USA
| |
Collapse
|
22
|
Mercer EM, Arrieta MC. Probiotics to improve the gut microbiome in premature infants: are we there yet? Gut Microbes 2023; 15:2201160. [PMID: 37122152 PMCID: PMC10153018 DOI: 10.1080/19490976.2023.2201160] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/06/2023] [Indexed: 05/02/2023] Open
Abstract
Gut microbiome maturation in infants born prematurely is uniquely influenced by the physiological, clinical, and environmental factors surrounding preterm birth and early life, leading to altered patterns of microbial succession relative to term infants during the first months of life. These differences in microbiome composition are implicated in acute clinical conditions that disproportionately affect preterm infants, including necrotizing enterocolitis (NEC) and late-onset sepsis (LOS). Probiotic supplementation initiated early in life is an effective prophylactic measure for preventing NEC, LOS, and other clinical concerns relevant to preterm infants. In parallel, reported benefits of probiotics on the preterm gut microbiome, metabolome, and immune function are beginning to emerge. This review summarizes the current literature on the influence of probiotics on the gut microbiome of preterm infants, outlines potential mechanisms by which these effects are exerted, and highlights important clinical considerations for determining the best practices for probiotic use in premature infants.
Collapse
Affiliation(s)
- Emily M. Mercer
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
- International Microbiome Center, University of Calgary, Calgary, Alberta, Canada
| | - Marie-Claire Arrieta
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
- International Microbiome Center, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
23
|
Mukhopadhyay S, Lee JJ, Hartman E, Woodford E, Dhudasia MB, Mattei LM, Daniel SG, Wade KC, Underwood MA, Bittinger K. Preterm infants at low risk for early-onset sepsis differ in early fecal microbiome assembly. Gut Microbes 2022; 14:2154091. [PMID: 36474348 PMCID: PMC9733690 DOI: 10.1080/19490976.2022.2154091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Antibiotics are administered near-universally to very low birth weight (VLBW) infants after birth for suspected early-onset sepsis (EOS). We previously identified a phenotypic group of VLBW infants, referred to as low-risk for EOS (LRE), whose risk of EOS is low enough to avoid routine antibiotic initiation. In this cohort study, we compared 18 such infants with 30 infants categorized as non-LRE to determine if the lower risk of pathogen transmission at birth is accompanied by differences in microbiome acquisition and development. We did shotgun metagenomic sequencing of 361 fecal samples obtained serially. LRE infants had a higher human-to-bacterial DNA ratio than non-LRE infants in fecal samples on days 1-3 after birth, confirming lower bacterial acquisition among LRE infants. The microbial diversity and composition in samples from days 4-7 differed between the groups with a predominance of Staphylococcus epidermidis in LRE infants and Enterobacteriaceae sp. in non-LRE infants. Compositional differences were congruent with the distribution of virulence factors and antibiotic resistant genes. After the first week, the overall composition was similar, but changes in relative abundance for several taxa with increasing age differed between groups. Of the nine late-onset bacteremia episodes, eight occurred in non-LRE infants. Species isolated from the blood culture was detected in the pre-antibiotic fecal samples of the infant for all episodes, though these species were also found in infants without bacteremia. In conclusion, LRE infants present a distinct pattern of microbiome development that is aligned with their low risk for EOS. Further investigation to determine the impact of these differences on later outcomes such as late-onset bacteremia is warranted.
Collapse
Affiliation(s)
- Sagori Mukhopadhyay
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States,Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States,Center for Pediatric Clinical Effectiveness, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States,Sagori Mukhopadhyay Center for Pediatric Clinical Effectiveness, Children’s Hospital of Philadelphia, Roberts Center for Pediatric Research, 2716 South Street, Office 19-322, Philadelphia, PA19146, United States
| | - Jung-Jin Lee
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
| | - Erica Hartman
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States,Center for Pediatric Clinical Effectiveness, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
| | - Emily Woodford
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
| | - Miren B. Dhudasia
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States,Center for Pediatric Clinical Effectiveness, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
| | - Lisa M. Mattei
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
| | - Scott G. Daniel
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
| | - Kelly C. Wade
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States,Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States
| | - Mark A. Underwood
- Department of Pediatrics, University of California Davis, Sacramento, California, United States
| | - Kyle Bittinger
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States,Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States,CONTACT Kyle Bittinger CHOP Microbiome Center, Children’s Hospital of Philadelphia, Roberts Center for Pediatric Research, 2716 South Street, Philadelphia, PA19146, United States
| |
Collapse
|
24
|
Ye Y, Forsythe S, Xu Z. Editorial: Infant food and intestinal immunity. Front Nutr 2022; 9:1089890. [PMID: 36505266 PMCID: PMC9732656 DOI: 10.3389/fnut.2022.1089890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 11/14/2022] [Indexed: 11/26/2022] Open
Affiliation(s)
- Yingwang Ye
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | | | - Zhenbo Xu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| |
Collapse
|
25
|
Vongbhavit K, Salinero LK, Kalanetra KM, Masarweh C, Yu A, Taft DH, Mills DA, Underwood MA. A comparison of bacterial colonization between nasogastric and orogastric enteral feeding tubes in infants in the neonatal intensive care unit. J Perinatol 2022; 42:1446-1452. [PMID: 35840710 PMCID: PMC9616717 DOI: 10.1038/s41372-022-01452-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 06/03/2022] [Accepted: 06/27/2022] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Feeding tubes harbor microbial contaminants; studies to date have not explored differences between orogastric (OG) and nasogastric (NG) tube biofilms. We sought to extend a previous analysis by comparing bacterial colonization by location (OG v NG) and by evaluating clinical factors that may affect tube bacterial populations. STUDY DESIGN The pharyngeal segments of 41 infant feeding tubes (14 OG and 27 NG) from 41 infants were analyzed by next generation 16 S rRNA sequencing on the MiSeq platform. RESULTS At the phylum level, Proteobacteria had the highest relative abundance of both OG and NG tubes. At the genus/species level, nine taxa differed significantly between OG and NG tubes. Alpha and beta diversity analyses showed significant differences between OG and NG tubes with relatively little contribution from clinical factors. CONCLUSION The route of feeding tube insertion (oral vs nasal) had a greater impact on bacterial colonization than the assessed clinical factors.
Collapse
Affiliation(s)
- Kannikar Vongbhavit
- Department of Pediatrics, HRH Princess Maha Chakri Sirindhorn Medical Center, Srinakharinwirot University, Nakornayok, Thailand
- Division of Neonatology, Department of Pediatrics, University of California Davis, Sacramento, CA, USA
| | - Lauren K Salinero
- Department of Food Science and Technology, University of California Davis, Davis, CA, USA
| | - Karen M Kalanetra
- Department of Food Science and Technology, University of California Davis, Davis, CA, USA
| | - Chad Masarweh
- Department of Food Science and Technology, University of California Davis, Davis, CA, USA
| | - Alice Yu
- Department of Food Science and Technology, University of California Davis, Davis, CA, USA
| | - Diana H Taft
- Department of Food Science and Technology, University of California Davis, Davis, CA, USA
| | - David A Mills
- Department of Food Science and Technology, University of California Davis, Davis, CA, USA
| | - Mark A Underwood
- Division of Neonatology, Department of Pediatrics, University of California Davis, Sacramento, CA, USA.
| |
Collapse
|
26
|
Casirati A, Somaschini A, Perrone M, Vandoni G, Sebastiani F, Montagna E, Somaschini M, Caccialanza R. Preterm birth and metabolic implications on later life: A narrative review focused on body composition. Front Nutr 2022; 9:978271. [PMID: 36185669 PMCID: PMC9521164 DOI: 10.3389/fnut.2022.978271] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 08/25/2022] [Indexed: 11/13/2022] Open
Abstract
Preterm newborn infants are characterized by low body weight and lower fat mass at birth compared with full-term newborn neonates. Conversely, at term corrected age, body fat mass is more represented in preterm newborn infants, causing a predisposition to developing metabolic syndrome and cardiovascular diseases in later life with a different risk profile in men as compared with women. Postnatal growth is a complex change in anthropometric parameters and body composition. Both quantity and quality of growth are regulated by several factors such as fetal programming, early nutrition, and gut microbiota. Weight gain alone is not an optimal indicator of nutritional status as it does not accurately describe weight quality. The analysis of body composition represents a potentially useful tool to predict later metabolic and cardiovascular risk as it detects the quality of growth by differentiating between fat and lean mass. Longitudinal follow-up of preterm newborn infants could take advantage of body composition analysis in order to identify high-risk patients who apply early preventive strategies. This narrative review aimed to examine the state-of-the-art body composition among born preterm children, with a focus on those in the pre-school age group.
Collapse
Affiliation(s)
- Amanda Casirati
- Clinical Nutrition and Dietetics Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- *Correspondence: Amanda Casirati,
| | - Alberto Somaschini
- Division of Cardiology and Cardiac Intensive Care Unit, San Paolo Hospital, Savona, Italy
| | - Michela Perrone
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Giulia Vandoni
- Clinical Nutrition, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Federica Sebastiani
- Endocrinology and Metabolic Diseases, Azienda USL IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Elisabetta Montagna
- Clinical Nutrition and Dietetics Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | | - Riccardo Caccialanza
- Clinical Nutrition and Dietetics Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
27
|
Liu M, Chen C, Kang S, Kwon JI, Jin J, Che H. Effect of different feeding methods and gut microbiota on premature infants and clinical outcomes. Front Nutr 2022; 9:888304. [PMID: 35978959 PMCID: PMC9376281 DOI: 10.3389/fnut.2022.888304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 07/06/2022] [Indexed: 11/20/2022] Open
Abstract
Premature infants require special care, and clinical feeding methods for this patient group are generally divided into breastfeeding and formula milk. This retrospective study investigated the effects of these two feeding methods on premature infants admitted to the neonatal intensive care unit between 2017 and 2018. Data regarding the duration of complete enteral feeding, weight gain, and postnatal infections were collected, categorized, and compared. Pearson's correlation coefficient was used to determine the correlation between the intestinal flora and clinical outcomes. Results revealed no differences between the two feeding methods, and neither had significant effects on clinical indicators in premature infants, although the gut microbiota may be an important factor influencing many clinical indicators. Results of this study suggest an important role for the gut microbiota in the care of premature infants and provide a basis for promoting the healthy development of this patient population.
Collapse
Affiliation(s)
- Manman Liu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Cheng Chen
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Songhao Kang
- College of Engineering, China Agricultural University, Beijing, China
| | - Jung-il Kwon
- Maeil Innovation Center, Maeil Dairies Co., Ltd., Seoul, South Korea
| | - Juan Jin
- Maeil Innovation Center, Maeil Dairies Co., Ltd., Seoul, South Korea
| | - Huilian Che
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| |
Collapse
|
28
|
Ma B, Sundararajan S, Nadimpalli G, France M, McComb E, Rutt L, Lemme-Dumit JM, Janofsky E, Roskes LS, Gajer P, Fu L, Yang H, Humphrys M, Tallon LJ, Sadzewicz L, Pasetti MF, Ravel J, Viscardi RM. Highly Specialized Carbohydrate Metabolism Capability in Bifidobacterium Strains Associated with Intestinal Barrier Maturation in Early Preterm Infants. mBio 2022; 13:e0129922. [PMID: 35695455 PMCID: PMC9239261 DOI: 10.1128/mbio.01299-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 05/11/2022] [Indexed: 12/26/2022] Open
Abstract
"Leaky gut," or high intestinal barrier permeability, is common in preterm newborns. The role of the microbiota in this process remains largely uncharacterized. We employed both short- and long-read sequencing of the 16S rRNA gene and metagenomes to characterize the intestinal microbiome of a longitudinal cohort of 113 preterm infants born between 240/7 and 326/7 weeks of gestation. Enabled by enhanced taxonomic resolution, we found that a significantly increased abundance of Bifidobacterium breve and a diet rich in mother's breastmilk were associated with intestinal barrier maturation during the first week of life. We combined these factors using genome-resolved metagenomics and identified a highly specialized genetic capability of the Bifidobacterium strains to assimilate human milk oligosaccharides and host-derived glycoproteins. Our study proposes mechanistic roles of breastmilk feeding and intestinal microbial colonization in postnatal intestinal barrier maturation; these observations are critical toward advancing therapeutics to prevent and treat hyperpermeable gut-associated conditions, including necrotizing enterocolitis (NEC). IMPORTANCE Despite improvements in neonatal intensive care, necrotizing enterocolitis (NEC) remains a leading cause of morbidity and mortality. "Leaky gut," or intestinal barrier immaturity with elevated intestinal permeability, is the proximate cause of susceptibility to NEC. Early detection and intervention to prevent leaky gut in "at-risk" preterm neonates are critical for decreasing the risk of potentially life-threatening complications like NEC. However, the complex interactions between the developing gut microbial community, nutrition, and intestinal barrier function remain largely uncharacterized. In this study, we reveal the critical role of a sufficient breastmilk feeding volume and the specialized carbohydrate metabolism capability of Bifidobacterium in the coordinated postnatal improvement of the intestinal barrier. Determining the clinical and microbial biomarkers that drive the intestinal developmental disparity will inform early detection and novel therapeutic strategies to promote appropriate intestinal barrier maturation and prevent NEC and other adverse health conditions in preterm infants.
Collapse
Affiliation(s)
- Bing Ma
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Sripriya Sundararajan
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Gita Nadimpalli
- Department of Epidemiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Michael France
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Elias McComb
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Lindsay Rutt
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jose M. Lemme-Dumit
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Elise Janofsky
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Lisa S. Roskes
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Pawel Gajer
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Li Fu
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Hongqiu Yang
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Mike Humphrys
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Luke J. Tallon
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Lisa Sadzewicz
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Marcela F. Pasetti
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jacques Ravel
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Rose M. Viscardi
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
29
|
Zhou L, Zeng Y, Zhang H, Ma Y. The Role of Gastrointestinal Microbiota in Functional Dyspepsia: A Review. Front Physiol 2022; 13:910568. [PMID: 35755434 PMCID: PMC9214042 DOI: 10.3389/fphys.2022.910568] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/23/2022] [Indexed: 12/18/2022] Open
Abstract
Functional dyspepsia is a clinically common functional gastrointestinal disorder with a high prevalence, high impact and high consumption of medical resources. The microbiota in the gastrointestinal tract is a large number of families and is one of the most complex microbial reservoirs in the human body. An increasing number of studies have confirmed the close association between dysbiosis of the gastrointestinal microbiota and the occurrence and progression of functional dyspepsia. Therefore, we reviewed the role of dysbiosis of the gastrointestinal microbiota, H. pylori infection and gastrointestinal microbiota metabolites in functional dyspepsia, focusing on the possible mechanisms by which dysbiosis of the gastrointestinal microbiota contributes to the pathogenesis of functional dyspepsia. Several studies have confirmed that dysbiosis of the gastrointestinal microbiota may cause the occurrence and progression of functional dyspepsia by disrupting the biological barrier of the intestinal mucosa, by disturbing the immune function of the intestinal mucosa, or by causing dysregulation of the microbial-gut-brain axis. Probiotics and antibiotics have also been chosen to treat functional dyspepsia in clinical studies and have shown some improvement in the clinical symptoms. However, more studies are needed to explore and confirm the relationship between dysbiosis of the gastrointestinal microbiota and the occurrence and progression of functional dyspepsia, and more clinical studies are needed to confirm the therapeutic efficacy of microbiota modulation for functional dyspepsia.
Collapse
Affiliation(s)
- Li Zhou
- Department of Rehabilitation Medicine, Wuhan Hospital of Integrated Traditional Chinese and Western Medicine, Wuhan, China
| | - Yi Zeng
- Department of Hospital Infection Management Office, Wuhan Hospital of Integrated Traditional Chinese and Western Medicine, Wuhan, China
| | - Hongxing Zhang
- Department of Acupuncture, Wuhan Hospital of Integrated Traditional Chinese and Western Medicine, Wuhan, China
| | - Yan Ma
- Department of Rehabilitation Medicine, Wuhan Hospital of Integrated Traditional Chinese and Western Medicine, Wuhan, China
| |
Collapse
|
30
|
Kaelin EA, Rodriguez C, Hall-Moore C, Hoffmann JA, Linneman LA, Ndao IM, Warner BB, Tarr PI, Holtz LR, Lim ES. Longitudinal gut virome analysis identifies specific viral signatures that precede necrotizing enterocolitis onset in preterm infants. Nat Microbiol 2022; 7:653-662. [PMID: 35449461 PMCID: PMC9064801 DOI: 10.1038/s41564-022-01096-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 03/02/2022] [Indexed: 12/22/2022]
Abstract
Necrotizing enterocolitis (NEC) is a serious consequence of preterm birth and is often associated with gut bacterial microbiome alterations. However, little is known about the development of the gut virome in preterm infants, or its role in NEC. Here, using metagenomic sequencing, we characterized the DNA gut virome of 9 preterm infants who developed NEC and 14 gestational age-matched preterm infants who did not. Infants were sampled longitudinally before NEC onset over the first 11 weeks of life. We observed substantial interindividual variation in the gut virome between unrelated preterm infants, while intraindividual variation over time was significantly less. We identified viral and bacterial signatures in the gut that preceded NEC onset. Specifically, we observed a convergence towards reduced viral beta diversity over the 10 d before NEC onset, which was driven by specific viral signatures and accompanied by specific viral-bacterial interactions. Our results indicate that bacterial and viral perturbations precede the sudden onset of NEC. These findings suggest that early life virome signatures in preterm infants may be implicated in NEC.
Collapse
Affiliation(s)
- Emily A Kaelin
- School of Life Sciences, Arizona State University, Tempe, AZ, USA.,Center for Fundamental and Applied Microbiomics, Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Cynthia Rodriguez
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO, USA
| | - Carla Hall-Moore
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO, USA
| | - Julie A Hoffmann
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO, USA
| | - Laura A Linneman
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO, USA
| | - I Malick Ndao
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO, USA
| | - Barbara B Warner
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO, USA
| | - Phillip I Tarr
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO, USA.,Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA
| | - Lori R Holtz
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO, USA.
| | - Efrem S Lim
- School of Life Sciences, Arizona State University, Tempe, AZ, USA. .,Center for Fundamental and Applied Microbiomics, Biodesign Institute, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
31
|
McDavid A, Laniewski N, Grier A, Gill AL, Kessler HA, Huyck H, Carbonell E, Holden-Wiltse J, Bandyopadhyay S, Carnahan J, Dylag AM, Topham DJ, Falsey AR, Caserta MT, Pryhuber GS, Gill SR, Scheible KM. Aberrant newborn T cell and microbiota developmental trajectories predict respiratory compromise during infancy. iScience 2022; 25:104007. [PMID: 35310935 PMCID: PMC8931366 DOI: 10.1016/j.isci.2022.104007] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/30/2021] [Accepted: 02/25/2022] [Indexed: 11/11/2022] Open
Abstract
Neonatal immune-microbiota co-development is poorly understood, yet age-appropriate recognition of - and response to - pathogens and commensal microbiota is critical to health. In this longitudinal study of 148 preterm and 119 full-term infants from birth through one year of age, we found that postmenstrual age or weeks from conception is a central factor influencing T cell and mucosal microbiota development. Numerous features of the T cell and microbiota functional development remain unexplained; however, by either age metric and are instead shaped by discrete perinatal and postnatal events. Most strikingly, we establish that prenatal antibiotics or infection disrupt the normal T cell population developmental trajectory, influencing subsequent respiratory microbial colonization and predicting respiratory morbidity. In this way, early exposures predict the postnatal immune-microbiota axis trajectory, placing infants at later risk for respiratory morbidity in early childhood.
Collapse
Affiliation(s)
- Andrew McDavid
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY, USA
| | - Nathan Laniewski
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | - Alex Grier
- Genomics Research Center, University of Rochester, Rochester, NY, USA
| | - Ann L. Gill
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | - Haeja A. Kessler
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | - Heidie Huyck
- Department of Pediatrics, University of Rochester, Rochester, NY, USA
| | | | - Jeanne Holden-Wiltse
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY, USA
| | - Sanjukta Bandyopadhyay
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY, USA
| | - Jennifer Carnahan
- Department of Pediatrics, University of Rochester, Rochester, NY, USA
| | - Andrew M. Dylag
- Department of Pediatrics, University of Rochester, Rochester, NY, USA
| | - David J. Topham
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | - Ann R. Falsey
- Department of Medicine, University of Rochester, Rochester, NY, USA
| | - Mary T. Caserta
- Department of Pediatrics, University of Rochester, Rochester, NY, USA
| | | | - Steven R. Gill
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | | |
Collapse
|
32
|
Capin I, Hinds A, Vomero B, Roth P, Blau J. Are Early-Onset Sepsis Evaluations and Empiric Antibiotics Mandatory for All Neonates Admitted with Respiratory Distress? Am J Perinatol 2022; 39:444-448. [PMID: 32947642 DOI: 10.1055/s-0040-1717070] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
OBJECTIVE The objective of this study was to evaluate the success and safety of an antimicrobial stewardship protocol for neonates admitted with respiratory distress at birth. STUDY DESIGN A retrospective cohort analysis of all infants admitted to the neonatal intensive care unit (NICU) with respiratory distress from January 2013 to February 2018 was conducted. In April 2016, an antimicrobial stewardship protocol was implemented, dividing neonates into two groups: maternal indications for delivery (no infectious risk factors for early-onset sepsis [EOS]) and fetal indications (risk factors present) for delivery. Neonates with risk factors for EOS were started on empiric antibiotics, those who lacked risk factors were observed. Paired sample t-test and descriptive statistics were used to compare the pre- and postprotocol implementation. RESULTS There were no missed cases of EOS in our study. Management with empiric antibiotics decreased from 95 to 41% of neonates with respiratory distress after initiation of the protocol. Newborns with a lower mean (±standard errors of the mean [SEM]) gestational age were more likely to receive empiric antibiotics (35.1 ± 0.4 [range: 23-42 weeks] vs. 37.7 ± 0.2 weeks [range: 24-42 weeks]; p < 0.05). Similar findings were seen for neonates with lower mean birth weights (2,627 ± 77 [range: 390-5,440 g] vs. 3,078 ± 51 g [range: 620-6,260 g]; p < 0.05). CONCLUSION The antibiotic stewardship protocol safely reduces the administration of empiric antibiotics to symptomatic neonates without missing any cases of sepsis. KEY POINTS · Newborns born with respiratory distress often receive broad-spectrum antibiotics upon NICU admission.. · An antibiotic stewardship program was created for this population and considered perinatal risk factors for sepsis when determining whether antibiotics were indicated.. · This antibiotic stewardship program was safe and effective, significantly reducing antibiotic use without missing any cases of sepsis..
Collapse
Affiliation(s)
- Ivana Capin
- Department of Pediatrics, Division of Neonatology, Staten Island University Hospital, Northwell Health, Staten Island, New York
| | - Autumn Hinds
- SUNY Downstate College of Medicine, Brooklyn, New York
| | - Bridgit Vomero
- Department of Pediatrics, Division of Neonatology, Staten Island University Hospital, Northwell Health, Staten Island, New York
| | - Philip Roth
- Department of Pediatrics, Division of Neonatology, Staten Island University Hospital, Northwell Health, Staten Island, New York.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
| | - Jonathan Blau
- Department of Pediatrics, Division of Neonatology, Staten Island University Hospital, Northwell Health, Staten Island, New York.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
| |
Collapse
|
33
|
Lemme-Dumit JM, Song Y, Lwin HW, Hernandez-Chavez C, Sundararajan S, Viscardi RM, Ravel J, Pasetti MF, Ma B. Altered Gut Microbiome and Fecal Immune Phenotype in Early Preterm Infants With Leaky Gut. Front Immunol 2022; 13:815046. [PMID: 35280991 PMCID: PMC8905226 DOI: 10.3389/fimmu.2022.815046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Intestinal barrier immaturity, or "leaky gut", is the proximate cause of susceptibility to necrotizing enterocolitis in preterm neonates. Exacerbated intestinal immune responses, gut microbiota dysbiosis, and heightened barrier injury are considered primary triggers of aberrant intestinal maturation in early life. Inordinate host immunity contributes to this process, but the precise elements remain largely uncharacterized, leaving a significant knowledge gap in the biological underpinnings of gut maturation. In this study, we investigated the fecal cytokine profile and gut microbiota in a cohort of 40 early preterm infants <33-weeks-gestation to identify immune markers of intestinal barrier maturation. Three distinct microbiota types were demonstrated to be differentially associated with intestinal permeability (IP), maternal breast milk feeding, and immunological profiles. The Staphylococcus epidermidis- and Enterobacteriaceae-predominant microbiota types were associated with an elevated IP, reduced breast milk feeding, and less defined fecal cytokine profile. On the other hand, a lower IP was associated with increased levels of fecal IL-1α/β and a microbiota type that included a wide array of anaerobes with expanded fermentative capacity. Our study demonstrated the critical role of both immunological and microbiological factors in the early development of intestinal barrier that collectively shape the intestinal microenvironment influencing gut homeostasis and postnatal intestinal maturation in early preterm newborns.
Collapse
Affiliation(s)
- Jose M Lemme-Dumit
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Yang Song
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Hnin Wai Lwin
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Claudia Hernandez-Chavez
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Sripriya Sundararajan
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Rose M Viscardi
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Jacques Ravel
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Marcela F Pasetti
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Bing Ma
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
34
|
Liu XC, Sun Q, Ji YC, Fu LZ, Wang ZL, He Y, Li LQ. Differences in the Gut Microbiota Composition and Metabolites Associated With Feeding Intolerance in VLBW Infants With a Gestational Age of ≤ 30 Weeks: A Pilot Study. Front Cell Infect Microbiol 2022; 12:726322. [PMID: 35252022 PMCID: PMC8891543 DOI: 10.3389/fcimb.2022.726322] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 01/27/2022] [Indexed: 12/12/2022] Open
Abstract
Objective To explore the main variations in gut microbiota compositions, short-chain fatty acids (SCFAs) concentrations and autoinducer-2 (AI-2) levels in very-low-birth-weight (VLBW) infants with feeding intolerance (FI). Methods Twenty-seven VLBW infants with gestational ages of ≤30 weeks were divided into the FI group (n=14) and feeding tolerance (FT) group (n=13). The gut microbiota composition and SCFAs concentrations and AI-2 levels in feces were detected at 2 and 4 weeks after birth. Results There was no difference in alpha diversity between the two groups at 2 and 4 weeks after birth (P>0.05). Although the Chao index decreased (P<0.05), there was no difference in the Shannon index from 2 weeks to 4 weeks in either the FI or FT group (P>0.05). Additionally, there was no difference in beta diversity between the FI and FT groups at 2 weeks (P>0.05), but there was a significant difference in beta diversity between the two groups at 4 weeks (P<0.05) and a large difference from 2 weeks to 4 weeks in both the FI and FT groups (P<0.05). Furthermore, the composition of the microbiota at 4 weeks was significantly different from that at 2 weeks in the FI group (P<0.05). The Veillonella abundance was lower at 4 weeks in the FI group (P<0.05), but there were no differences in the compositions of the other main microbes between the two groups (P>0.05). Proteobacteria and Firmicutes were dominant in both the FI and FT groups. The concentrations of propanoic, valeric and hexanoic acids were lower in the FI group at 2 weeks, and the levels of isobutyric and valeric acids were lower at 4 weeks after birth (P<0.05). The areas under the curves (AUCs) of propanoic, butanoic and valeric acids in predicting FI were 0.878, 0.816 and 0.744, respectively. Compared with that in the FT group, the relative bioluminescence of AI-2 was lower in the FI group at 2 weeks (P<0.05), and the AUC was 0.736. Conclusions The main composition of the microbiota was not obviously different in infants with FI. Some SCFAs and AI-2 have moderate value in predicting FI.
Collapse
Affiliation(s)
- Xiao-Chen Liu
- Neonatal Diagnosis and Treatment Centre of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Qian Sun
- Neonatal Diagnosis and Treatment Centre of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Yan-Chun Ji
- Neonatal Diagnosis and Treatment Centre of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Li-Zhen Fu
- Neonatal Diagnosis and Treatment Centre of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Zheng-Li Wang
- Neonatal Diagnosis and Treatment Centre of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Yu He
- Neonatal Diagnosis and Treatment Centre of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Lu-Quan Li
- Neonatal Diagnosis and Treatment Centre of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
- *Correspondence: Lu-Quan Li,
| |
Collapse
|
35
|
The Microbiota-Gut Axis in Premature Infants: Physio-Pathological Implications. Cells 2022; 11:cells11030379. [PMID: 35159189 PMCID: PMC8834399 DOI: 10.3390/cells11030379] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/22/2021] [Accepted: 01/17/2022] [Indexed: 11/16/2022] Open
Abstract
Intriguing evidence is emerging in regard to the influence of gut microbiota composition and function on host health from the very early stages of life. The development of the saprophytic microflora is conditioned by several factors in infants, and peculiarities have been found for babies born prematurely. This population is particularly exposed to a high risk of infection, postnatal antibiotic treatment, feeding difficulties and neurodevelopmental disabilities. To date, there is still a wide gap in understanding all the determinants and the mechanism behind microbiota disruption and its influence in the development of the most common complications of premature infants. A large body of evidence has emerged during the last decades showing the existence of a bidirectional communication axis involving the gut microbiota, the gut and the brain, defined as the microbiota–gut–brain axis. In this context, given that very few data are available to demonstrate the correlation between microbiota dysbiosis and neurodevelopmental disorders in preterm infants, increasing interest has arisen to better understand the impact of the microbiota–gut–brain axis on the clinical outcomes of premature infants and to clarify how this may lead to alternative preventive, diagnostic and therapeutic strategies. In this review, we explored the current evidence regarding microbiota development in premature infants, focusing on the effects of delivery mode, type of feeding, environmental factors and possible influence of the microbiota–gut–brain axis on preterm clinical outcomes during their hospital stay and on their health status later in life.
Collapse
|
36
|
Sprenger N, Tytgat HL, Binia A, Austin S, Singhal A. Biology of human milk oligosaccharides: from Basic Science to Clinical Evidence. J Hum Nutr Diet 2022; 35:280-299. [PMID: 35040200 PMCID: PMC9304252 DOI: 10.1111/jhn.12990] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 01/05/2022] [Indexed: 11/28/2022]
Abstract
Human milk oligosaccharides (HMOs) have been researched by scientists for over 100 years, driven by the substantial evidence for the nutritional and health benefits of mother's milk. Yet research has truly bloomed during the last decade, thanks to the progress in biotechnology, which allowed the production of large amounts of bona fide HMOs. The availability of HMOs has been particularly crucial for the renewed interest in HMO research because of the low abundance or even absence of HMOs in farmed animal milk. This interest is reflected in the increasing number of original research publications and reviews on HMOs. Here, we provide an overview and critical discussion on structure function relations of HMOs that highlight why they are such interesting and important components of human milk. Clinical observations in breastfed infants backed by basic research from animal models provide guidance as to what physiological roles for HMOs are to be expected. From an evidence-based nutrition viewpoint, we discuss the current data supporting clinical relevance of specific HMOs based on randomized placebo controlled clinical intervention trials in formula-fed infants. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Norbert Sprenger
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Hanne Lp Tytgat
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Aristea Binia
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Sean Austin
- Nestlé Institute of Food Safety and Analytical Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Atul Singhal
- Institute of Child Health, University College London, London, WC1N 1EH, United Kingdom
| |
Collapse
|
37
|
Brunse A, Offersen SM, Mosegaard JJ, Deng L, Damborg P, Nielsen DS, Sangild PT, Thymann T, Nguyen DN. Enteral broad-spectrum antibiotics antagonize the effect of fecal microbiota transplantation in preterm pigs. Gut Microbes 2022; 13:1-16. [PMID: 33382952 PMCID: PMC7781584 DOI: 10.1080/19490976.2020.1849997] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
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.
Collapse
Affiliation(s)
- Anders Brunse
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Simone Margaard Offersen
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Josefine Juliane Mosegaard
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ling Deng
- Section of Food Microbiology and Fermentation, Department of Food Science, Faculty of Science, Copenhagen University, Copenhagen, Denmark
| | - Peter Damborg
- Section for Veterinary Clinical Microbiology, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Dennis Sandris Nielsen
- Section of Food Microbiology and Fermentation, Department of Food Science, Faculty of Science, Copenhagen University, Copenhagen, Denmark
| | - Per Torp Sangild
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Thymann
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Duc Ninh Nguyen
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark,CONTACT Duc Ninh Nguyen Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
38
|
Aguilar-Lopez M, Dinsmoor AM, Ho TTB, Donovan SM. A systematic review of the factors influencing microbial colonization of the preterm infant gut. Gut Microbes 2022; 13:1-33. [PMID: 33818293 PMCID: PMC8023245 DOI: 10.1080/19490976.2021.1884514] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Prematurity coupled with the necessary clinical management of preterm (PT) infants introduces multiple factors that can interfere with microbial colonization. This study aimed to review the perinatal, physiological, pharmacological, dietary, and environmental factors associated with gut microbiota of PT infants. A total of 587 articles were retrieved from a search of multiple databases. Sixty studies were included in the review after removing duplicates and articles that did not meet the inclusion criteria. Review of this literature revealed that evidence converged on the effect of postnatal age, mode of delivery, use of antibiotics, and consumption of human milk in the composition of gut microbiota of PT infants. Less evidence was found for associations with race, sex, use of different fortifiers, macronutrients, and other medications. Future studies with rich metadata are needed to further explore the impact of the PT exposome on the development of the microbiota in this high-risk population.
Collapse
Affiliation(s)
- Miriam Aguilar-Lopez
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, USA
| | - Andrew M. Dinsmoor
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, USA
| | - Thao T. B. Ho
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, USA
| | - Sharon M. Donovan
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, USA,Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, USA,CONTACT Sharon M. Donovan Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, 339 Bevier Hall 905 S. Goodwin Avenue, Urbana, IL61801, USA
| |
Collapse
|
39
|
Socha-Banasiak A, Pawłowska M, Czkwianianc E, Pierzynowska K. From Intrauterine to Extrauterine Life-The Role of Endogenous and Exogenous Factors in the Regulation of the Intestinal Microbiota Community and Gut Maturation in Early Life. Front Nutr 2022; 8:696966. [PMID: 34977104 PMCID: PMC8718557 DOI: 10.3389/fnut.2021.696966] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
Differentiation of the digestive tube and formation of the gut unit as a whole, are regulated by environmental factors through epigenetic modifications which enhance cellular plasticity. The critical period of DNA imprinting lasts from conception until approximately the 1,000th day of human life. During pregnancy, besides agents that may directly promote epigenetic programming (e.g., folate, zinc, and choline supplementation), some factors (e.g., antibiotic use, dietary components) can affect the composition of the mother's microbiota, in turn affecting the fetal microbiome which interacts with the offspring's intestinal epithelial cells. According to available literature that confirms intrauterine microbial colonization, the impact of the microbiome and its metabolites on the genome seems to be key in fetal development, including functional gut maturation and the general health status of the offspring, as well as later on in life. Although the origin of the fetal microbiome is still not well-understood, the bacteria may originate from both the vagina, as the baby is born, as well as from the maternal oral cavity/gut, through the bloodstream. Moreover, the composition of the fetal gut microbiota varies depending on gestational age, which in turn possibly affects the regulation of the immune system at the barrier between mother and fetus, leading to differences in the ability of microorganisms to access and survive in the fetal environment. One of the most important local functions of the gut microbiota during the prenatal period is their exposure to foreign antigens which in turn contributes to immune system and tissue development, including fetal intestinal Innate Lymphoid Cells (ILCs). Additional factors that determine further infant microbiome development include whether the infant is born premature or at term, the method of delivery, maternal antibiotic use, and the composition of the mother's milk, among others. However, the latest findings highlight the fact that a more diverse infant gut microbiome at birth facilitates the proliferation of stem cells by microbial metabolites and accelerates infant development. This phenomenon confirms the unique role of microbiome. This review emphasizes the crucial perinatal and postnatal factors that may influence fetal and neonatal microbiota, and in turn gut maturation.
Collapse
Affiliation(s)
- Anna Socha-Banasiak
- Department of Gastroenterology, Allergology and Pediatrics, Polish Mother's Memorial Hospital-Research Institute, Lodz, Poland
| | - Malwina Pawłowska
- Department of Gastroenterology, Allergology and Pediatrics, Polish Mother's Memorial Hospital-Research Institute, Lodz, Poland
| | - Elżbieta Czkwianianc
- Department of Gastroenterology, Allergology and Pediatrics, Polish Mother's Memorial Hospital-Research Institute, Lodz, Poland
| | - Kateryna Pierzynowska
- Department of Biology, Lund University, Lund, Sweden.,Department of Animal Physiology, The Kielanowski Institute of Animal Nutrition and Physiology Polish Academy of Sciences, Jablonna, Poland
| |
Collapse
|
40
|
Huang YE, Shen X, Yin D, Lan S, Lu Y, Zhou P, Ma L, Zhang Y, Sheng Y, Zhang Y, Li M, Hu F, Chen J, Li P, El-Omar EM, Zheng H. Disrupted establishment of anaerobe and facultative anaerobe balance in preterm infants with extrauterine growth restriction. Front Pediatr 2022; 10:935458. [PMID: 36147811 PMCID: PMC9486202 DOI: 10.3389/fped.2022.935458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 08/08/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Extrauterine growth restriction (EUGR) in preterm birth infants could have long-term adverse impacts on health. Less is known about the gut microbiota regarding its establishment in early life and its role in long-term growth in preterm birth infants. METHODS A prospective, longitudinal observational study was conducted with 67 preterm infants in a level III neonatal intensive care unit. Clinical information was obtained from medical records, and fecal samples were collected weekly during hospitalization and processed for 16S rRNA gene sequencing. RESULTS The bacterial profiles from the weekly sampling of preterm infants demonstrated that the early-life gut microbiota was clustered into the following four stages in chronological order: stage 1: 0-4 days, stage 2: 1-2 weeks, stage 3: 3-7 weeks, and stage 4: 8-10 weeks. The development of gut microbiota showed latency at stage 4 in EUGR infants compared with that in non-EUGR infants, which resulted from their consistently high level of facultative anaerobes, including Enterobacteriaceae and Staphylococcus, and lack of obligate anaerobes, including Clostridium and Veillonella. In the 2-year follow-up, infants with a high level of obligate anaerobes-to-facultative anaerobes ratio at stage 4 had a lower risk of long-term growth restriction at the margin of statistical significance. CONCLUSION The results of this study indicate that the development of gut microbiota in the early life of EUGR infants is delayed compared with that of non-EUGR infants. The obligate-to-facultative anaerobes ratio could be an indicator of the maturity of gut microbiota development and associated with the risk of long-term growth restriction in preterm infants.
Collapse
Affiliation(s)
- Yi-E Huang
- Department of Hospital Infection Control, Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen, China
| | - Xintian Shen
- Department of Pharmacy, Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen, China
| | - Dingding Yin
- Department of Neonatology, Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen, China
| | - Shanwei Lan
- Department of Obstetrics and Gynecology, The First People's Hospital of Foshan, Foshan, China.,The Second Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Yongxue Lu
- Department of Neonatology, The First People's Hospital of Foshan, Foshan, China
| | - Ping Zhou
- Department of Neonatology, Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen, China
| | - Liya Ma
- Department of Neonatology, Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen, China
| | - Yinlan Zhang
- Department of Hospital Infection Control, Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen, China
| | - Yuhui Sheng
- Department of Hospital Infection Control, Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen, China
| | - Youjun Zhang
- Department of Hospital Infection Control, Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen, China
| | - Mengna Li
- Department of Neonatology, Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen, China
| | - Fei Hu
- Department of Neonatology, Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen, China
| | - Jiaqi Chen
- Department of Neonatology, Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen, China
| | - Pan Li
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campuses, UNSW Sydney, Sydney, NSW, Australia
| | - Emad M El-Omar
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campuses, UNSW Sydney, Sydney, NSW, Australia
| | - Huimin Zheng
- Department of Obstetrics and Gynecology, The First People's Hospital of Foshan, Foshan, China.,UNSW Microbiome Research Centre, St George and Sutherland Clinical Campuses, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
41
|
The gut microbiome of extremely preterm infants randomized to the early progression of enteral feeding. Pediatr Res 2022; 92:799-804. [PMID: 34775476 PMCID: PMC9098696 DOI: 10.1038/s41390-021-01831-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 09/07/2021] [Accepted: 10/09/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Early progression of feeding could influence the development of the gut microbiome. METHODS We collected fecal samples from extremely preterm infants randomized to receive either early (feeding day 2) or delayed (feeding day 5) feeding progression. After study completion, we compared samples obtained at three different time points (week 1, week 2, and week 3) to determine longitudinal differences in specific taxa between the study groups using unadjusted and adjusted negative binomial and zero-inflated mixed models. Analyses were adjusted for a mode of delivery, breastmilk intake, and exposure to antibiotics. RESULTS We analyzed 137 fecal samples from 51 infants. In unadjusted and adjusted analyses, we did not observe an early transition to higher microbial diversity within samples (i.e., alpha diversity) or significant differences in microbial diversity between samples (i.e., beta diversity) in the early feeding group. Our longitudinal, single-taxon analysis found consistent differences in the genera Lactococcus, Veillonella, and Bilophila between groups. CONCLUSIONS Differences in single-taxon analyses independent of the mode of delivery, exposure to antibiotics, and breastmilk feeding suggest potential benefits of early progression of enteral feeding volumes. However, this dietary intervention does not appear to increase the diversity of the gut microbiome in the first 28 days after birth. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT02915549. IMPACT Early progression of enteral feeding volumes with human milk reduces the duration of parenteral nutrition and the need for central venous access among extremely preterm infants. Early progression of enteral feeding leads to single-taxon differences in longitudinal analyses of the gut microbiome, but it does not appear to increase the diversity of the gut microbiome in the first 28 days after birth. Randomization in enteral feeding trials creates appealing opportunities to evaluate the effects of human milk diets on the gut microbiome.
Collapse
|
42
|
Romano-Keeler J, Sun J. The First 1000 Days: Assembly of the Neonatal Microbiome and Its Impact on Health Outcomes. NEWBORN (CLARKSVILLE, MD.) 2022; 1:219-226. [PMID: 36237439 PMCID: PMC9555117 DOI: 10.5005/jp-journals-11002-0028] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Early life microbial colonization is critical for the development of the immune system, postnatal growth, and long-term health and disease. The dynamic and nascent microbiomes of children are highly individualized and are characterized by low bacterial diversity. Any disruptions in microbial colonization can contribute to shifts in normal microbial colonization that persist past the first 1000 days of life and result in intestinal dysbiosis. Here, we focus on microbiome-host interactions during fetal, newborn, and infant microbiome development. We summarize the roles of bacterial communities in fetal development and adverse health outcomes due to dysbiosis. We also discuss how internal and external factors program the microbiome's metabolic machinery as it evolves into an adult-like microbiome. Finally, we discuss the limits of current studies and future directions. Studies on the early-life microbiome will be critical for a better understanding of childhood health and diseases, as well as restorative methods for the prevention and treatment of diseases in adulthood.
Collapse
Affiliation(s)
- Joann Romano-Keeler
- Division of Neonatology, Department of Pediatrics, University of Illinois, Chicago, Illinois, United States of America
| | - Jun Sun
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois, Chicago, Illinois, United States of America; University of Illinois Cancer Center, Chicago, Illinois, United States of America; Jesse Brown VA Medical Center, Chicago, Illinois, United States of America
| |
Collapse
|
43
|
Luo S, Zhu H, Zhang J, Wan D. The Pivotal Role of Microbiota in Modulating the Neuronal-Glial-Epithelial Unit. Infect Drug Resist 2021; 14:5613-5628. [PMID: 34992388 PMCID: PMC8711043 DOI: 10.2147/idr.s342782] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/26/2021] [Indexed: 12/12/2022] Open
Abstract
The enteric nervous system (ENS) consists of enteric neurons and enteric glial cells (EGCs) and controls the function of the epithelial barrier. Thus, a novel concept of neuronal-glial-epithelial unit in the gut was put forward by analogy with neuronal-glial-endothelial unit in the brain. The environment in the gastrointestinal (GI) tract is complex as it harbours millions of bacteria, which extensively attach with intestinal epithelium. The cross-talk between the neuronal-glial-endothelial unit and microbiota plays a pivotal role in modulating the epithelial barrier's permeability, intestinal development and immune response. And evidence shows dysbiosis is the potent risk factor in the pathologic process of Parkinson's disease (PD) and inflammatory bowel disease (IBD). In this review, we summarize the compelling results in favor of microbiota serving as the key modulator in the neuronal-glial-epithelial unit development and function, with profound effects on intestinal homeostasis.
Collapse
Affiliation(s)
- Siyu Luo
- Department of Emergency & Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Huifeng Zhu
- College of Pharmaceutical Sciences & Chinese Medicine, Southwest University, Chongqing, People’s Republic of China
| | - Junhui Zhang
- Health Management Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Dong Wan
- Department of Emergency & Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| |
Collapse
|
44
|
|
45
|
Fonseca W, Malinczak CA, Fujimura K, Li D, McCauley K, Li J, Best SKK, Zhu D, Rasky AJ, Johnson CC, Bermick J, Zoratti EM, Ownby D, Lynch SV, Lukacs NW, Ptaschinski C. Maternal gut microbiome regulates immunity to RSV infection in offspring. J Exp Med 2021; 218:212680. [PMID: 34613328 PMCID: PMC8500238 DOI: 10.1084/jem.20210235] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/26/2021] [Accepted: 09/07/2021] [Indexed: 12/15/2022] Open
Abstract
Development of the immune system can be influenced by diverse extrinsic and intrinsic factors that influence the risk of disease. Severe early life respiratory syncytial virus (RSV) infection is associated with persistent immune alterations. Previously, our group had shown that adult mice orally supplemented with Lactobacillus johnsonii exhibited decreased airway immunopathology following RSV infection. Here, we demonstrate that offspring of mice supplemented with L. johnsonii exhibit reduced airway mucus and Th2 cell–mediated response to RSV infection. Maternal supplementation resulted in a consistent gut microbiome in mothers and their offspring. Importantly, supplemented maternal plasma and breastmilk, and offspring plasma, exhibited decreased inflammatory metabolites. Cross-fostering studies showed that prenatal Lactobacillus exposure led to decreased Th2 cytokines and lung inflammation following RSV infection, while postnatal Lactobacillus exposure diminished goblet cell hypertrophy and mucus production in the lung in response to airway infection. These studies demonstrate that Lactobacillus modulation of the maternal microbiome and associated metabolic reprogramming enhance airway protection against RSV in neonates.
Collapse
Affiliation(s)
- Wendy Fonseca
- Department of Pathology, University of Michigan, Ann Arbor, MI
| | | | - Kei Fujimura
- Department of Medicine-Gastroenterology, University of California, San Francisco, San Francisco, CA
| | - Danny Li
- Department of Medicine-Gastroenterology, University of California, San Francisco, San Francisco, CA
| | - Kathryn McCauley
- Department of Medicine-Gastroenterology, University of California, San Francisco, San Francisco, CA
| | - Jia Li
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI
| | | | - Diana Zhu
- Department of Pathology, University of Michigan, Ann Arbor, MI
| | - Andrew J Rasky
- Department of Pathology, University of Michigan, Ann Arbor, MI
| | | | - Jennifer Bermick
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Michigan, Ann Arbor, MI
| | - Edward M Zoratti
- Division of Allergy and Clinical Immunology, Department of Medicine, Henry Ford Health System, Detroit, MI
| | - Dennis Ownby
- Department of Pediatrics, Augusta University, Augusta, GA
| | - Susan V Lynch
- Department of Medicine-Gastroenterology, University of California, San Francisco, San Francisco, CA
| | - Nicholas W Lukacs
- Department of Pathology, University of Michigan, Ann Arbor, MI.,Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI
| | - Catherine Ptaschinski
- Department of Pathology, University of Michigan, Ann Arbor, MI.,Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI
| |
Collapse
|
46
|
Medise BE. Growth and Development in Preterm Infants: What is The Long-Term Risk? AMERTA NUTRITION 2021. [DOI: 10.20473/amnt.v5i1sp.2021.27-33] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
ABSTRACT Background: Indonesia comes in the fifth for the greatest number of preterm births. Preterm infants may inflict various complication as the result of underdeveloped immunity, affecting their growth and development in the long run until they reach adult phase. Such complications could be prevented through adequate nutrition fulfillment. Purpose: This article aimed to elaborate the characteristics of growth and development of premature babies, long term effect on the development and the impact of immunity and gut health of preterm infants in supporting their growth and development. Methods: References cited in this article were obtained from the latest primary literature within the last 10 years. Discussion: The rate and ability of infants to perform catch-up growth depends on the birth weight and gestation age, at which the lower birth weight and lower gestational age had slower rate. Brain structures that of preterm infants differ compared to the term, and these changes give rise to various clinical outcomes, including long term emotional, behavioral changes, cognitive and executive functioning. Immature immune system in preterm infants reduces the protective ability by innate and adaptive immunity in overcoming pathogens compared to term infants, including gut microbiota prematurity which affects nutrition absorption and growth and development catch up ability. Appropriate and adequate nutrition supplementation has shown beneficial effects in promoting the growth of normal gut flora, which allow better absorption of nutrition and therefore enhancing growth rate and supporting the development of preterm infants. Conclusions: Optimal growth and development of preterm infants are supported by sufficient nutrition supplementation to support the growth of gut microbiota, facilitating the catch-up growth and development of premature infants and immune system maturity.
Collapse
|
47
|
From Mum to Bum: An Observational Study Protocol to Follow Digestion of Human Milk Oligosaccharides and Glycoproteins from Mother to Preterm Infant. Nutrients 2021; 13:nu13103430. [PMID: 34684428 PMCID: PMC8538091 DOI: 10.3390/nu13103430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/21/2021] [Accepted: 09/24/2021] [Indexed: 11/21/2022] Open
Abstract
The nutritional requirements of preterm infants are challenging to meet in neonatal care, yet crucial for their growth, development and health. Aberrant maturation of the gastrointestinal tract and the microbiota could affect the digestion of human milk and its nutritional value considerably. Therefore, the main objective of the proposed research is to investigate how the intestinal microbiota of preterm and full-term infants differ in their ability to extract energy and nutrients from oligosaccharides and glycoproteins in human milk. This pilot study will be an observational, single-center study performed at the Neonatal Intensive Care Unit at Isala Women and Children’s Hospital (Zwolle, The Netherlands). A cohort of thirty mother–infant pairs (preterm ≤30 weeks of gestation, n = 15; full-term 37–42 weeks of gestation, n = 15) will be followed during the first six postnatal weeks with follow-up at three- and six-months postnatal age. We will collect human milk of all mothers, gastric aspirates of preterm infants and fecal samples of all infants. A combination of 16S rRNA amplicon sequencing, proteomics, peptidomics, carbohydrate analysis and calorimetric measurements will be performed. The role of the microbiota in infant growth and development is often overlooked yet offers opportunities to advance neonatal care. The ‘From Mum to Bum’ study is the first study in which the effect of a preterm gut microbiota composition on its metabolic capacity and subsequent infant growth and development is investigated. By collecting human milk of all mothers, gastric aspirates of preterm infants and fecal samples of all infants at each timepoint, we can follow digestion of human milk from the breast of the mother throughout the gastrointestinal tract of the infant, or ‘From Mum to Bum’.
Collapse
|
48
|
Wu TT, Xiao J, Sohn MB, Fiscella KA, Gilbert C, Grier A, Gill AL, Gill SR. Machine Learning Approach Identified Multi-Platform Factors for Caries Prediction in Child-Mother Dyads. Front Cell Infect Microbiol 2021; 11:727630. [PMID: 34490147 PMCID: PMC8417465 DOI: 10.3389/fcimb.2021.727630] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 07/09/2021] [Indexed: 12/19/2022] Open
Abstract
Untreated tooth decays affect nearly one third of the world and is the most prevalent disease burden among children. The disease progression of tooth decay is multifactorial and involves a prolonged decrease in pH, resulting in the demineralization of tooth surfaces. Bacterial species that are capable of fermenting carbohydrates contribute to the demineralization process by the production of organic acids. The combined use of machine learning and 16s rRNA sequencing offers the potential to predict tooth decay by identifying the bacterial community that is present in an individual’s oral cavity. A few recent studies have demonstrated machine learning predictive modeling using 16s rRNA sequencing of oral samples, but they lack consideration of the multifactorial nature of tooth decay, as well as the role of fungal species within their models. Here, the oral microbiome of mother–child dyads (both healthy and caries-active) was used in combination with demographic–environmental factors and relevant fungal information to create a multifactorial machine learning model based on the LASSO-penalized logistic regression. For the children, not only were several bacterial species found to be caries-associated (Prevotella histicola, Streptococcus mutans, and Rothia muciloginosa) but also Candida detection and lower toothbrushing frequency were also caries-associated. Mothers enrolled in this study had a higher detection of S. mutans and Candida and a higher plaque index. This proof-of-concept study demonstrates the significant impact machine learning could have in prevention and diagnostic advancements for tooth decay, as well as the importance of considering fungal and demographic–environmental factors.
Collapse
Affiliation(s)
- Tong Tong Wu
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, United States
| | - Jin Xiao
- Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, NY, United States
| | - Michael B Sohn
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, United States
| | - Kevin A Fiscella
- Department of Family Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - Christie Gilbert
- Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - Alex Grier
- Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - Ann L Gill
- Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - Steve R Gill
- Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
49
|
Shen W, Qiu W, Liu Y, Liao W, Ma Y, He Y, Wang Z, Zhou H. Postnatal age is strongly correlated with the early development of the gut microbiome in preterm infants. Transl Pediatr 2021; 10:2313-2324. [PMID: 34733672 PMCID: PMC8506066 DOI: 10.21037/tp-21-367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/27/2021] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The gut microbiome plays a potential role in clinical events in preterm infants and may affect their lateral development. Understanding the initial colonization of microbes in the gut, their early dynamic changes, and the major factors correlated with these changes would provide crucial information about the developmental process in early life. METHODS The present study enrolled 151 preterm infants and examined the longitudinal dynamics of their fecal microbiome profiles during the period of hospitalization using 16S ribosomal RNA gene sequencing. Random forest modeling was used to predict postnatal age (Age), postmenstrual age (PMA), and gestational age (GA), using gut microbiome features. RESULTS Principal coordinate analysis revealed that the gut microbiome of the preterm infants displayed an obvious time-dependent change pattern, which showed the strongest association with Age, followed by PMA, and a much weaker association with (GA). Random forest modeling further evidenced the time-dependent change pattern, with the Pearson's correlation coefficients between the actual values and the gut microbiome-predicted values being 0.68, 0.53, and 0.38 for postnatal, postmenstrual, and gestational age, respectively. The microbiome dynamism could be further divided into four Age stages, each with its own characteristic microbial taxa. The first 1-4 days (T1 stage) represented the meconium microbiome, with colonization of a high diversity of microbes before or during delivery. During 5-15 days (T2 stage), the gut microbiome of the preterm infants underwent a rapid turnover, in which microbial diversity declined, and stabilized afterward. Enterobacteriaceae, Enterococcaceae, Streptococcaceae, Staphylococcaceae, and Clostridiaceae were the major classes in the gut microbiome in the lateral stages of development (T3-T4 stage). CONCLUSIONS Postnatal age, rather than the gestational age, is significantly correlated with the gut microbiome of preterm infants, suggesting that clinical interventions contribute more to the early dynamics of gut microbiome in preterm infants than the natural development of the gut.
Collapse
Affiliation(s)
- Wei Shen
- Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wen Qiu
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yuting Liu
- Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weihua Liao
- Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yiyi Ma
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yan He
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zhang Wang
- Institute of Ecological Sciences, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Hongwei Zhou
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
50
|
Yang K, He S, Dong W. Gut microbiota and bronchopulmonary dysplasia. Pediatr Pulmonol 2021; 56:2460-2470. [PMID: 34077996 DOI: 10.1002/ppul.25508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/02/2021] [Accepted: 05/16/2021] [Indexed: 12/20/2022]
Abstract
Bronchopulmonary dysplasia is a relatively common and severe complication of prematurity, and its pathogenesis remains ambiguous. Revolutionary advances in microbiological analysis techniques, together with the growing sophistication of the gut-lung axis hypothesis, have resulted in more studies linking gut microbiota dysbiosis to the occurrence and development of bronchopulmonary dysplasia. The present article builds on current findings to examine the intrinsic associations between gut microbiota and bronchopulmonary dysplasia. Gut microbiota dysbiosis may insult the intestinal barrier, triggering inflammation, metabolic disturbances, and malnutrition, consequences of which might impact bronchopulmonary dysplasia by altering the gut-lung axis. By evaluating the potential mechanisms, new therapeutic targets and potential therapeutic modalities for bronchopulmonary dysplasia can be identified from a microecological perspective.
Collapse
Affiliation(s)
- Kun Yang
- Department of Pediatrics, Division of Neonatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Shasha He
- Department of Pediatrics, Division of Neonatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wenbin Dong
- Department of Pediatrics, Division of Neonatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|